1
|
Shukla M, Narayan M. Proteostasis and Its Role in Disease Development. Cell Biochem Biophys 2025; 83:1725-1741. [PMID: 39422790 PMCID: PMC12123047 DOI: 10.1007/s12013-024-01581-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/22/2024] [Indexed: 10/19/2024]
Abstract
Proteostasis (protein homeostasis) refers to the general biological process that maintains the proper balance between the synthesis of proteins, their folding, trafficking, and degradation. It ensures proteins are functional, locally distributed, and appropriately folded inside cells. Genetic information enclosed in mRNA is translated into proteins. To ensure newly synthesized proteins take on the exact three-dimensional conformation, molecular chaperones assist in proper folding. Misfolded proteins can be refolded or targeted for elimination to stop aggregation. Cells utilize different degradation pathways, for instance, the ubiquitin-proteasome system, the autophagy-lysosome pathway, and the unfolded protein response, to degrade unwanted or damaged proteins. Quality control systems of the cell monitor the folding of proteins. These checkpoint mechanisms are aimed at degrading or refolding misfolded or damaged proteins. Under stress response pathways, such as heat shock response and unfolded protein response, which are triggered under conditions that perturb proteostasis, the capacity for folding is increased, and degradation pathways are activated to help cells handle stressful conditions. The deregulation of proteostasis is implicated in a variety of illnesses, comprising cancer, metabolic diseases, cardiovascular diseases, and neurological disorders. Therapeutic strategies with a deeper insight into the mechanism of proteostasis are crucial for the treatment of illnesses linked with proteostasis and to support cellular health. Thus, proteostasis is required not only for the maintenance of cellular homeostasis and function but also for proper protein function and prevention of injurious protein aggregation. In this review, we have covered the concept of proteostasis, its mechanism, and how disruptions to it can result in a number of disorders.
Collapse
Affiliation(s)
- Manisha Shukla
- Department of Biotechnology, Pandit S.N. Shukla University, Shahdol, Madhya Pradesh, India
| | - Mahesh Narayan
- Department of Chemistry and Biochemistry, University of Texas, El Paso, TX, USA.
| |
Collapse
|
2
|
Peltier S, Marin M, Dzieciatkowska M, Dussiot M, Roy MK, Bruce J, Leblanc L, Hadjou Y, Georgeault S, Fricot A, Roussel C, Stephenson D, Casimir M, Sissoko A, Paye F, Dokmak S, Ndour PA, Roingeard P, Gautier EF, Spitalnik SL, Hermine O, Buffet PA, D’Alessandro A, Amireault P. Proteostasis and metabolic dysfunction characterize a subset of storage-induced senescent erythrocytes targeted for posttransfusion clearance. J Clin Invest 2025; 135:e183099. [PMID: 40067362 PMCID: PMC12043093 DOI: 10.1172/jci183099] [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: 06/11/2024] [Accepted: 03/05/2025] [Indexed: 03/19/2025] Open
Abstract
Although refrigerated storage slows the metabolism of volunteer donor RBCs, which is essential in transfusion medicine, cellular aging still occurs throughout this in vitro process. Storage-induced microerythrocytes (SMEs) are morphologically altered senescent RBCs that accumulate during storage and are cleared from circulation following transfusion. However, the molecular and cellular alterations that trigger clearance of this RBC subset remain to be identified. Using a staining protocol that sorts long-stored SMEs (i.e., CFSEhi) and morphologically normal RBCs (CFSElo), these in vitro aged cells were characterized. Metabolomics analysis identified depletion of energy, lipid-repair, and antioxidant metabolites in CFSEhi RBCs. By redox proteomics, irreversible protein oxidation primarily affected CFSEhi RBCs. By proteomics, 96 proteins, mostly in the proteostasis family, had relocated to CFSEhi RBC membranes. CFSEhi RBCs exhibited decreased proteasome activity and deformability; increased phosphatidylserine exposure, osmotic fragility, and endothelial cell adherence; and were cleared from the circulation during human spleen perfusion ex vivo. Conversely, molecular, cellular, and circulatory properties of long-stored CFSElo RBCs resembled those of short-stored RBCs. CFSEhi RBCs are morphologically and metabolically altered, have irreversibly oxidized and membrane-relocated proteins, and exhibit decreased proteasome activity. In vitro aging during storage selectively alters metabolism and proteostasis in these storage-induced senescent RBCs targeted for clearance.
Collapse
Affiliation(s)
- Sandy Peltier
- Université Paris Cité, INSERM, BIGR, Paris, France
- Université Paris Cité, Institut Imagine, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM, Paris, France
| | | | - Monika Dzieciatkowska
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Michaël Dussiot
- Université Paris Cité, Institut Imagine, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM, Paris, France
| | - Micaela Kalani Roy
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Johanna Bruce
- Proteom’IC facility, Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | | | | | - Sonia Georgeault
- Plateforme des Microscopies, Infrastructures de Recherche en Biologie Santé et Agronomie, Programme Pluriformation Analyse des Systèmes Biologiques, Tours, France
| | | | - Camille Roussel
- Université Paris Cité, INSERM, BIGR, Paris, France
- Laboratoire d’hématologie générale, Hôpital Universitaire Necker Enfants Malades, Assistance Publique des Hôpitaux de Paris (AP-HP), Paris, France
| | - Daniel Stephenson
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Madeleine Casimir
- Université Paris Cité, Institut Imagine, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM, Paris, France
- Université Libre de Bruxelles, Hôpital Erasme, Département d’Hématologie, Brussels, Belgium
| | | | - François Paye
- Department of Digestive Surgery, Hôpital Saint-Antoine, AP-HP, Paris, France
| | - Safi Dokmak
- Department of Hepatobiliary Surgery and Liver Transplantation, Hôpital Beaujon, AP-HP, Clichy, France
| | | | - Philippe Roingeard
- Plateforme des Microscopies, Infrastructures de Recherche en Biologie Santé et Agronomie, Programme Pluriformation Analyse des Systèmes Biologiques, Tours, France
- U1259, Morphogenèse et Antigénicité du VIH et des Virus des Hépatites, INSERM, Université de Tours and Centre Hospitalier Régional Universitaire de Tours, Tours, France
| | - Emilie-Fleur Gautier
- Proteom’IC facility, Université Paris Cité, CNRS, INSERM, Institut Cochin, Paris, France
| | - Steven L. Spitalnik
- Department of Pathology and Cell Biology, Columbia University, New York, New York, USA
| | - Olivier Hermine
- Université Paris Cité, Institut Imagine, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM, Paris, France
- Département d’Hématologie, Hôpital Universitaire Necker Enfants Malades, AP-HP, Paris, France
| | - Pierre A. Buffet
- Université Paris Cité, INSERM, BIGR, Paris, France
- Service des maladies infectieuses et tropicales, Hôpital Universitaire Necker Enfants Malades, AP-HP, Paris, France
| | - Angelo D’Alessandro
- Department of Biochemistry and Molecular Genetics, University of Colorado Denver, Anschutz Medical Campus, Aurora, Colorado, USA
| | - Pascal Amireault
- Université Paris Cité, INSERM, BIGR, Paris, France
- Université Paris Cité, Institut Imagine, Laboratory of Cellular and Molecular Mechanisms of Hematological Disorders and Therapeutic Implications, INSERM, Paris, France
| |
Collapse
|
3
|
Hudák A, Letoha T. Endocytic Pathways Unveil the Role of Syndecans in the Seeding and Spreading of Pathological Protein Aggregates: Insights into Neurodegenerative Disorders. Int J Mol Sci 2025; 26:4037. [PMID: 40362276 PMCID: PMC12071627 DOI: 10.3390/ijms26094037] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2025] [Revised: 04/07/2025] [Accepted: 04/23/2025] [Indexed: 05/15/2025] Open
Abstract
Alzheimer's disease and other neurodegenerative disorders are characterized by the accumulation of misfolded proteins, such as amyloid-beta, tau, and α-synuclein, which disrupt neuronal function and contribute to cognitive decline. Heparan sulfate proteoglycans, particularly syndecans, play a pivotal role in the seeding, aggregation, and spreading of toxic protein aggregates through endocytic pathways. Among these, syndecan-3 is particularly critical in regulating the internalization of misfolded proteins, facilitating their propagation in a prion-like manner. This review examines the mechanisms by which syndecans, especially SDC3, contribute to the seeding and spreading of pathological protein aggregates in neurodegenerative diseases. Understanding these endocytic pathways provides valuable insights into the potential of syndecans as biomarkers and therapeutic targets for early intervention in Alzheimer's disease and other related neurodegenerative disorders.
Collapse
Affiliation(s)
- Anett Hudák
- Pharmacoidea Ltd., 6726 Szeged, Hungary;
- Doctoral School of Theoretical Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| | - Tamás Letoha
- Pharmacoidea Ltd., 6726 Szeged, Hungary;
- Doctoral School of Theoretical Medicine, Albert Szent-Györgyi Medical School, University of Szeged, 6720 Szeged, Hungary
| |
Collapse
|
4
|
Salem S, Alpaugh M, Saint-Pierre M, Alves-Martins-Borba FN, Cerquera-Cleves C, Lemieux M, Ngonza-Nito SB, De Koninck P, Melki R, Cicchetti F. Treatment with Tau fibrils impact Huntington's disease-related phenotypes in cell and mouse models. Neurobiol Dis 2024; 202:106696. [PMID: 39389154 DOI: 10.1016/j.nbd.2024.106696] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2024] [Revised: 09/13/2024] [Accepted: 10/04/2024] [Indexed: 10/12/2024] Open
Abstract
There is now compelling evidence for the presence of pathological forms of Tau in tissues of both patients and animal models of Huntington's disease (HD). While the root cause of this illness is a mutation within the huntingtin gene, a number of studies now suggest that HD could also be considered a secondary tauopathy. However, the contributory role of Tau in the pathogenesis and pathophysiology of this condition, as well as its implications in cellular toxicity and consequent behavioral impairments are largely unknown. We therefore performed intracerebral stereotaxic injections of recombinant human Tau monomers and fibrils into the knock-in zQ175 mouse model of HD. Tau fibrils induced cognitive and anxiety-like phenotypes predominantly in zQ175 mice and increased the number and size of insoluble mutant huntingtin (mHTT) aggregates in the brains of treated animals. To better understand the putative mechanisms through which Tau could initiate and/or contribute to pathology, we incubated StHdh striatal cells, an in vitro model of HD, with the different Tau forms and evaluated the effects on cell functionality and heat shock proteins Hsp70 and Hsp90. Calcium imaging experiments showed functional impairments of HD StHdh cells following treatment with Tau fibrils, as well as significant changes to the levels of both heat shock proteins which were found trapped within mHTT aggregates. The accumulation of Hsp70 and 90 within aggregates was also present in mouse tissue which suggests that alteration of molecular chaperone-dependent protein quality control may influence aggregation, implicating proteostasis in the mHTT-Tau interplay.
Collapse
Affiliation(s)
- Shireen Salem
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada
| | - Melanie Alpaugh
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Martine Saint-Pierre
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada
| | - Flavia Natale Alves-Martins-Borba
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Catalina Cerquera-Cleves
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada
| | - Mado Lemieux
- CERVO Brain Research Center, 2601 de la Canardière, Québec, QC G1J 2G3, Canada
| | - Soki Bradel Ngonza-Nito
- Labortory of Neurodegenerative Diseases, Institut François Jacob, MIRCen, CEA, CNRS, Fontenay-aux-Roses, France
| | - Paul De Koninck
- CERVO Brain Research Center, 2601 de la Canardière, Québec, QC G1J 2G3, Canada
| | - Ronald Melki
- Labortory of Neurodegenerative Diseases, Institut François Jacob, MIRCen, CEA, CNRS, Fontenay-aux-Roses, France
| | - Francesca Cicchetti
- Cente de Recherche du CHU de Québec, Axe Neurosciences, T2-07, 2705, Boulevard Laurier, Québec, QC G1V 4G2, Canada; Département de Médecine Moléculaire, Université Laval, Québec, QC, Canada; Département de Psychiatrie et Neurosciences, Université Laval, Québec, QC, Canada.
| |
Collapse
|
5
|
Singh SB, Rajput SS, Sharma A, Kataria S, Dutta P, Ananthanarayanan V, Nandi A, Patil S, Majumdar A, Subramanyam D. Pathogenic Huntingtin aggregates alter actin organization and cellular stiffness resulting in stalled clathrin-mediated endocytosis. eLife 2024; 13:e98363. [PMID: 39382268 PMCID: PMC11643626 DOI: 10.7554/elife.98363] [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: 04/02/2024] [Accepted: 09/01/2024] [Indexed: 10/10/2024] Open
Abstract
Aggregation of mutant forms of Huntingtin is the underlying feature of neurodegeneration observed in Huntington's disorder. In addition to neurons, cellular processes in non-neuronal cell types are also shown to be affected. Cells expressing neurodegeneration-associated mutant proteins show altered uptake of ligands, suggestive of impaired endocytosis, in a manner as yet unknown. Using live cell imaging, we show that clathrin-mediated endocytosis (CME) is affected in Drosophila hemocytes and mammalian cells containing Huntingtin aggregates. This is also accompanied by alterations in the organization of the actin cytoskeleton resulting in increased cellular stiffness. Further, we find that Huntingtin aggregates sequester actin and actin-modifying proteins. Overexpression of Hip1 or Arp3 (actin-interacting proteins) could restore CME and cellular stiffness in cells containing Huntingtin aggregates. Neurodegeneration driven by pathogenic Huntingtin was also rescued upon overexpression of either Hip1 or Arp3 in Drosophila. Examination of other pathogenic aggregates revealed that TDP-43 also displayed defective CME, altered actin organization and increased stiffness, similar to pathogenic Huntingtin. Together, our results point to an intimate connection between dysfunctional CME, actin misorganization and increased cellular stiffness caused by alteration in the local intracellular environment by pathogenic aggregates.
Collapse
Affiliation(s)
- Surya Bansi Singh
- National Centre for Cell Science, SP Pune University CampusPuneIndia
- SP Pune UniversityPuneIndia
| | - Shatruhan Singh Rajput
- Indian Institute of Science Education and ResearchPuneIndia
- Department of Biochemistry, University of Cambridge, 80 Tennis Court RoadCambridgeUnited Kingdom
| | - Aditya Sharma
- Department of Computer Science and Engineering, Indian Institute of Technology Bombay, PowaiMumbaiIndia
| | - Sujal Kataria
- Indian Institute of Science Education and ResearchPuneIndia
| | - Priyanka Dutta
- National Centre for Cell Science, SP Pune University CampusPuneIndia
| | - Vaishnavi Ananthanarayanan
- EMBL Australia Node in Single Molecule Science, School of Biomedical Sciences, University of New South WalesSydneyAustralia
| | - Amitabha Nandi
- Department of Physics, Indian Institute of Technology, Bombay PowaiMumbaiIndia
| | | | - Amitabha Majumdar
- National Centre for Cell Science, SP Pune University CampusPuneIndia
| | - Deepa Subramanyam
- National Centre for Cell Science, SP Pune University CampusPuneIndia
| |
Collapse
|
6
|
Peltier S, Marin M, Dzieciatkowska M, Dussiot M, Roy MK, Bruce J, Leblanc L, Hadjou Y, Georgeault S, Fricot A, Roussel C, Stephenson D, Casimir M, Sissoko A, Paye F, Dokmak S, Ndour PA, Roingeard P, Gautier EF, Spitalnik SL, Hermine O, Buffet PA, D'Alessandro A, Amireault P. Proteostasis and metabolic dysfunction in a distinct subset of storage-induced senescent erythrocytes targeted for clearance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.11.612195. [PMID: 39314353 PMCID: PMC11419012 DOI: 10.1101/2024.09.11.612195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/25/2024]
Abstract
Although refrigerated storage slows the metabolism of volunteer donor RBCs, cellular aging still occurs throughout this in vitro process, which is essential in transfusion medicine. Storage-induced microerythrocytes (SMEs) are morphologically-altered senescent RBCs that accumulate during storage and which are cleared from circulation following transfusion. However, the molecular and cellular alterations that trigger clearance of this RBC subset remain to be identified. Using a staining protocol that sorts long-stored SMEs (i.e., CFSE high ) and morphologically-normal RBCs (CFSE low ), these in vitro aged cells were characterized. Metabolomics analysis identified depletion of energy, lipid-repair, and antioxidant metabolites in CFSE high RBCs. By redox proteomics, irreversible protein oxidation primarily affected CFSE high RBCs. By proteomics, 96 proteins, mostly in the proteostasis family, had relocated to CFSE high RBC membranes. CFSE high RBCs exhibited decreased proteasome activity and deformability; increased phosphatidylserine exposure, osmotic fragility, and endothelial cell adherence; and were cleared from the circulation during human spleen ex vivo perfusion. Conversely, molecular, cellular, and circulatory properties of long-stored CFSE low RBCs resembled those of short-stored RBCs. CFSE high RBCs are morphologically and metabolically altered, have irreversibly oxidized and membrane-relocated proteins, and exhibit decreased proteasome activity. In vitro aging during storage selectively alters metabolism and proteostasis in SMEs, targeting these senescent cells for clearance.
Collapse
|
7
|
Zhou X, Yang Y, Tai Z, Zhang H, Yang J, Luo Z, Xu Z. The mechanism of mitochondrial autophagy regulating Clathrin-mediated endocytosis in epilepsy. Epilepsia Open 2024; 9:1252-1264. [PMID: 38700951 PMCID: PMC11296089 DOI: 10.1002/epi4.12945] [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: 07/08/2023] [Revised: 03/10/2024] [Accepted: 03/31/2024] [Indexed: 05/05/2024] Open
Abstract
OBJECTIVE The objective of this study is to determine whether inhibition of mitophagy affects seizures through Clathrin-mediated endocytosis (CME). METHODS Pentylenetetrazol (PTZ) was intraperitoneally injected daily to establish a chronic PTZ-kindled seizure. The Western blot (WB) was used to compare the differences in Parkin protein expression between the epilepsy group and the control group. Immunofluorescence was used to detect the expression of MitoTracker and LysoTracker. Transferrin-Alexa488 (Tf-A488) was injected into the hippocampus of mice. We evaluated the effect of 3-methyladenine (3-MA) on epilepsy behavior through observation in PTZ-kindled models. RESULTS The methylated derivative of adenine, known as 3-MA, has been extensively utilized in the field of autophagy research. The transferrin protein is internalized from the extracellular environment into the intracellular space via the CME pathway. Tf-A488 uses a fluorescent marker to track CME. Western blot showed that the expression of Parkin was significantly increased in the PTZ-kindled model (p < 0.05), while 3-MA could reduce the expression (p < 0.05). The fluorescence uptake of MitoTracker and LysoTracker was increased in the primary cultured neurons induced by magnesium-free extracellular fluid (p < 0.05); the fluorescence uptake of Tf-A488 was significantly decreased in the 3-MA group compared with the control group (p < 0.05). Following hippocampal injection of Tf-A488, both the epilepsy group and the 3-MA group exhibited decreased fluorescence uptake, with a more pronounced effect observed in the 3-MA group. Inhibition of mitophagy by 3-MA from day 3 to day 9 progressively exacerbated seizure severity and shortened latency. SIGNIFICANCE It is speculated that the aggravation of seizures by 3-MA may be related to the failure to remove damaged mitochondria in time and effectively after inhibiting mitochondrial autophagy, affecting the vesicle endocytosis function of CME and increasing the susceptibility to epilepsy. SUMMARY Abnormal mitophagy was observed in a chronic pentylenetetrazol-induced seizure model and a Mg2+-free-induced spontaneous recurrent epileptiform discharge model. A fluorescent transferrin marker was utilized to track clathrin-mediated endocytosis. Using an autophagy inhibitor (3-methyladenine) on primary cultured neurons, we discovered that inhibition of autophagy led to a reduction in fluorescent transferrin uptake, while impairing clathrin-mediated endocytosis function mediated by mitophagy. Finally, we examined the effects of 3-methyladenine in an animal model of seizures showing that it exacerbated seizure severity. Ultimately, this study provides insights into potential mechanisms through which mitophagy regulates clathrin-mediated endocytosis in epilepsy.
Collapse
Affiliation(s)
- Xuejiao Zhou
- Department of NeurologyThe Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative Innovation Center of Tissue Damage Repair and RegenerationZunyi Medical UniversityZunyiChina
| | - Yu Yang
- Department of NeurologyThe Affiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Zhenzhen Tai
- Department of NeurologyThe Affiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Haiqing Zhang
- Department of NeurologyThe Affiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Juan Yang
- Department of NeurologyThe Affiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Zhong Luo
- Department of NeurologyThe Affiliated Hospital of Zunyi Medical UniversityZunyiChina
| | - Zucai Xu
- Department of NeurologyThe Affiliated Hospital of Zunyi Medical UniversityZunyiChina
- The Collaborative Innovation Center of Tissue Damage Repair and RegenerationZunyi Medical UniversityZunyiChina
- Key Laboratory of Brain ScienceZunyi Medical UniversityZunyiChina
| |
Collapse
|
8
|
Hipp MS, Hartl FU. Interplay of Proteostasis Capacity and Protein Aggregation: Implications for Cellular Function and Disease. J Mol Biol 2024; 436:168615. [PMID: 38759929 DOI: 10.1016/j.jmb.2024.168615] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2024] [Revised: 05/13/2024] [Accepted: 05/13/2024] [Indexed: 05/19/2024]
Abstract
Eukaryotic cells are equipped with an intricate proteostasis network (PN), comprising nearly 3,000 components dedicated to preserving proteome integrity and sustaining protein homeostasis. This protective system is particularly important under conditions of external and intrinsic cell stress, where inherently dynamic proteins may unfold and lose functionality. A decline in proteostasis capacity is associated with the aging process, resulting in a reduced folding efficiency of newly synthesized proteins and a deficit in the cellular capacity to degrade misfolded proteins. A critical consequence of PN insufficiency is the accumulation of cytotoxic protein aggregates that underlie various age-related neurodegenerative conditions and other pathologies. By interfering with specific proteostasis components, toxic aggregates place an excessive burden on the PN's ability to maintain proteome integrity. This initiates a feed-forward loop, wherein the generation of misfolded and aggregated proteins ultimately leads to proteostasis collapse and cellular demise.
Collapse
Affiliation(s)
- Mark S Hipp
- Department of Biomedical Sciences, University Medical Center Groningen, University of Groningen, Antonius Deusinglaan, 1, 9713 AV Groningen, the Netherlands; Research School of Behavioural and Cognitive Neurosciences, University of Groningen, Groningen, the Netherlands; School of Medicine and Health Sciences, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.
| | - F Ulrich Hartl
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany; Munich Cluster for Systems Neurology (SyNergy), Munich, Germany; Aligning Science Across Parkinson's (ASAP) Collaborative Research Network, Chevy Chase, MD, USA.
| |
Collapse
|
9
|
Ferrari L, Bauer B, Qiu Y, Schuschnig M, Klotz S, Anrather D, Juretschke T, Beli P, Gelpi E, Martens S. Tau fibrils evade autophagy by excessive p62 coating and TAX1BP1 exclusion. SCIENCE ADVANCES 2024; 10:eadm8449. [PMID: 38865459 PMCID: PMC11168460 DOI: 10.1126/sciadv.adm8449] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/09/2023] [Accepted: 05/07/2024] [Indexed: 06/14/2024]
Abstract
The accumulation of protein aggregates is a hallmark of many diseases, including Alzheimer's disease. As a major pillar of the proteostasis network, autophagy mediates the degradation of protein aggregates. The autophagy cargo receptor p62 recognizes ubiquitin on proteins and cooperates with TAX1BP1 to recruit the autophagy machinery. Paradoxically, protein aggregates are not degraded in various diseases despite p62 association. Here, we reconstituted the recognition by the autophagy receptors of physiological and pathological Tau forms. Monomeric Tau recruits p62 and TAX1BP1 via the sequential actions of the chaperone and ubiquitylation machineries. In contrast, Tau fibrils from Alzheimer's disease brains are recognized by p62 but fail to recruit TAX1BP1. This failure is due to the masking of fibrils ubiquitin moieties by p62. Tau fibrils are resistant to deubiquitylation, and, thus, this nonproductive interaction of p62 with the fibrils is irreversible. Our results shed light on the mechanism underlying autophagy evasion by protein aggregates and their consequent accumulation in disease.
Collapse
Affiliation(s)
- Luca Ferrari
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
- University of Vienna, Max Perutz Labs, Department of Biochemistry and Cell Biology, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
| | - Bernd Bauer
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
- University of Vienna, Max Perutz Labs, Department of Biochemistry and Cell Biology, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
- Vienna BioCenter PhD Program, Doctoral School of the University of Vienna and Medical University of Vienna, Campus-Vienna-Biocenter 1, 1030 Vienna, Austria
| | - Yue Qiu
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
- University of Vienna, Max Perutz Labs, Department of Biochemistry and Cell Biology, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
| | - Martina Schuschnig
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
- University of Vienna, Max Perutz Labs, Department of Biochemistry and Cell Biology, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
| | - Sigrid Klotz
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Dorothea Anrather
- Max Perutz Labs, Mass Spectrometry Facility, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
| | | | - Petra Beli
- Institute of Molecular Biology, 55128 Mainz, Germany
- Institute of Developmental Biology and Neurobiology (IDN), Johannes Gutenberg-Universität, 55128 Mainz, Germany
| | - Ellen Gelpi
- Division of Neuropathology and Neurochemistry, Department of Neurology, Medical University of Vienna, 1090 Vienna, Austria
| | - Sascha Martens
- Max Perutz Labs, Vienna Biocenter Campus (VBC), Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
- University of Vienna, Max Perutz Labs, Department of Biochemistry and Cell Biology, Dr.-Bohr-Gasse 9, 1030 Vienna, Austria
| |
Collapse
|
10
|
D'Urso B, Weil R, Génin P. [Optineurin and mitochondrial dysfunction in neurodegeneration]. Med Sci (Paris) 2024; 40:167-175. [PMID: 38411425 DOI: 10.1051/medsci/2023220] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/28/2024] Open
Abstract
Optineurin (OPTN) is a multifunctional protein playing a crucial role as a receptor in selective autophagy. OPTN gene mutations are linked to diseases such as normal-tension glaucoma and amyotrophic lateral sclerosis. Recognized as a critical receptor for mitophagy, OPTN is pivotal in selectively degrading damaged mitochondria. This process is essential to prevent their accumulation, the generation of reactive oxygen species, and the release of pro-apoptotic factors. Mitophagy's quality control is governed by the PINK1 kinase and the cytosolic ubiquitin ligase Parkin, whose mutations are associated with Parkinson's disease. This review highlights recent insights emphasizing OPTN's role in mitophagy and its potential involvement in neurodegenerative diseases.
Collapse
Affiliation(s)
- Baptiste D'Urso
- CIMI-Paris, UPMC UMRS CR7 - Inserm U1135 - CNRS EMR8255, Faculté de médecine Sorbonne Université site Pitié-Salpêtrière, Paris, France - Sorbonne Université, Faculté des sciences et ingénierie, Paris, France
| | - Robert Weil
- CIMI-Paris, UPMC UMRS CR7 - Inserm U1135 - CNRS EMR8255, Faculté de médecine Sorbonne Université site Pitié-Salpêtrière, Paris, France
| | - Pierre Génin
- CIMI-Paris, UPMC UMRS CR7 - Inserm U1135 - CNRS EMR8255, Faculté de médecine Sorbonne Université site Pitié-Salpêtrière, Paris, France
| |
Collapse
|
11
|
Stöberl N, Donaldson J, Binda CS, McAllister B, Hall-Roberts H, Jones L, Massey TH, Allen ND. Mutant huntingtin confers cell-autonomous phenotypes on Huntington's disease iPSC-derived microglia. Sci Rep 2023; 13:20477. [PMID: 37993517 PMCID: PMC10665390 DOI: 10.1038/s41598-023-46852-z] [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: 08/28/2023] [Accepted: 11/06/2023] [Indexed: 11/24/2023] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by a dominantly inherited CAG repeat expansion in the huntingtin gene (HTT). Neuroinflammation and microglia have been implicated in HD pathology, however it has been unclear if mutant HTT (mHTT) expression has an adverse cell-autonomous effect on microglial function, or if they are only activated in response to the neurodegenerative brain environment in HD. To establish a human cell model of HD microglia function, we generated isogenic controls for HD patient-derived induced pluripotent stem cells (iPSC) with 109 CAG repeats (Q109). Q109 and isogenic Q22 iPSC, as well as non-isogenic Q60 and Q33 iPSC lines, were differentiated to iPSC-microglia. Our study supports a model of basal microglia dysfunction in HD leading to elevated pro-inflammatory cytokine production together with impaired phagocytosis and endocytosis capacity, in the absence of immune stimulation. These findings are consistent with early microglia activation observed in pre-manifest patients and indicate that mHTT gene expression affects microglia function in a cell-autonomous way.
Collapse
Affiliation(s)
- Nina Stöberl
- School of Biosciences, Cardiff University, Cardiff, UK.
| | - Jasmine Donaldson
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Caroline S Binda
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Branduff McAllister
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Hazel Hall-Roberts
- UK Dementia Research Institute at Cardiff, Cardiff University, Cardiff, UK
| | - Lesley Jones
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | - Thomas H Massey
- Centre for Neuropsychiatric Genetics and Genomics, Division of Psychological Medicine and Clinical Neurosciences, Cardiff University, Cardiff, UK
| | | |
Collapse
|
12
|
Zhou S, Zhou Y, Zhong W, Su Z, Qin Z. Involvement of protein L-isoaspartyl methyltransferase in the physiopathology of neurodegenerative diseases: Possible substrates associated with synaptic function. Neurochem Int 2023; 170:105606. [PMID: 37657764 DOI: 10.1016/j.neuint.2023.105606] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2023] [Revised: 08/11/2023] [Accepted: 08/29/2023] [Indexed: 09/03/2023]
Abstract
Synaptic dysfunction is a typical pathophysiologic change in neurodegenerative diseases (NDs) such as Alzheimer's disease (AD), Parkinson's disease (PD), Hintington's disease (HD) and amyotrophic lateral sclerosis (ALS), which involves protein post-translational modifications (PTMs) including L-isoaspartate (L-isoAsp) formed by isomerization of aspartate or deamidation of asparagine. The formation of L-isoAsp could be repaired by protein L-isoaspartyl methyltransferase (PIMT). Some synaptic proteins have been identified as PIMT potential substrates and play an essential role in ensuring synaptic function. In this review, we discuss the role of certain synaptic proteins as PIMT substrates in neurodegenerative disease, thus providing therapeutic synapse-centered targets for the treatment of NDs.
Collapse
Affiliation(s)
- Sirui Zhou
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Yancheng Zhou
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Wanyu Zhong
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Zhonghao Su
- Department of Febrile Disease, School of Traditional Chinese Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| | - Zhenxia Qin
- Department of Anatomy, School of Integrative Medicine, Shanghai University of Traditional Chinese Medicine, Shanghai, China.
| |
Collapse
|
13
|
Pei Y, Lin C, Li H, Feng Z. Genetic background influences pig responses to porcine reproductive and respiratory syndrome virus. Front Vet Sci 2023; 10:1289570. [PMID: 37929286 PMCID: PMC10623566 DOI: 10.3389/fvets.2023.1289570] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 10/09/2023] [Indexed: 11/07/2023] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) is a highly infectious and economically significant virus that causes respiratory and reproductive diseases in pigs. It results in reduced productivity and increased mortality in pigs, causing substantial economic losses in the industry. Understanding the factors affecting pig responses to PRRSV is crucial to develop effective control strategies. Genetic background has emerged as a significant determinant of susceptibility and resistance to PRRSV in pigs. This review provides an overview of the basic infection process of PRRSV in pigs, associated symptoms, underlying immune mechanisms, and roles of noncoding RNA and alternative splicing in PRRSV infection. Moreover, it emphasized breed-specific variations in these aspects that may have implications for individual treatment options.
Collapse
Affiliation(s)
- Yangli Pei
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Key Laboratory of Animal Molecular Design and Precise Breeding of Guangdong Higher Education Institutes, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Chenghong Lin
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Key Laboratory of Animal Molecular Design and Precise Breeding of Guangdong Higher Education Institutes, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Hua Li
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Key Laboratory of Animal Molecular Design and Precise Breeding of Guangdong Higher Education Institutes, School of Life Science and Engineering, Foshan University, Foshan, China
| | - Zheng Feng
- Guangdong Provincial Key Laboratory of Animal Molecular Design and Precise Breeding, Key Laboratory of Animal Molecular Design and Precise Breeding of Guangdong Higher Education Institutes, School of Life Science and Engineering, Foshan University, Foshan, China
| |
Collapse
|
14
|
Quanrud GM, Lyu Z, Balamurugan SV, Canizal C, Wu HT, Genereux JC. Cellular Exposure to Chloroacetanilide Herbicides Induces Distinct Protein Destabilization Profiles. ACS Chem Biol 2023; 18:1661-1676. [PMID: 37427419 PMCID: PMC10367052 DOI: 10.1021/acschembio.3c00338] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2023] [Accepted: 06/23/2023] [Indexed: 07/11/2023]
Abstract
Herbicides in the widely used chloroacetanilide class harbor a potent electrophilic moiety, which can damage proteins through nucleophilic substitution. In general, damaged proteins are subject to misfolding. Accumulation of misfolded proteins compromises cellular integrity by disrupting cellular proteostasis networks, which can further destabilize the cellular proteome. While direct conjugation targets can be discovered through affinity-based protein profiling, there are few approaches to probe how cellular exposure to toxicants impacts the stability of the proteome. We apply a quantitative proteomics methodology to identify chloroacetanilide-destabilized proteins in HEK293T cells based on their binding to the H31Q mutant of the human Hsp40 chaperone DNAJB8. We find that a brief cellular exposure to the chloroacetanilides acetochlor, alachlor, and propachlor induces misfolding of dozens of cellular proteins. These herbicides feature distinct but overlapping profiles of protein destabilization, highly concentrated in proteins with reactive cysteine residues. Consistent with the recent literature from the pharmacology field, reactivity is driven by neither inherent nucleophilic nor electrophilic reactivity but is idiosyncratic. We discover that propachlor induces a general increase in protein aggregation and selectively targets GAPDH and PARK7, leading to a decrease in their cellular activities. Hsp40 affinity profiling identifies a majority of propachlor targets identified by competitive activity-based protein profiling (ABPP), but ABPP can only identify about 10% of protein targets identified by Hsp40 affinity profiling. GAPDH is primarily modified by the direct conjugation of propachlor at a catalytic cysteine residue, leading to global destabilization of the protein. The Hsp40 affinity strategy is an effective technique to profile cellular proteins that are destabilized by cellular toxin exposure. Raw proteomics data is available through the PRIDE Archive at PXD030635.
Collapse
Affiliation(s)
- Guy M. Quanrud
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Ziqi Lyu
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Sunil V. Balamurugan
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Carolina Canizal
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Hoi-Ting Wu
- Department of Chemistry, University of California, Riverside, California 92521, United States
| | - Joseph C. Genereux
- Department of Chemistry, University of California, Riverside, California 92521, United States
| |
Collapse
|
15
|
Kohler V, Andréasson C. Reversible protein assemblies in the proteostasis network in health and disease. Front Mol Biosci 2023; 10:1155521. [PMID: 37021114 PMCID: PMC10067754 DOI: 10.3389/fmolb.2023.1155521] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2023] [Accepted: 03/09/2023] [Indexed: 04/07/2023] Open
Abstract
While proteins populating their native conformations constitute the functional entities of cells, protein aggregates are traditionally associated with cellular dysfunction, stress and disease. During recent years, it has become clear that large aggregate-like protein condensates formed via liquid-liquid phase separation age into more solid aggregate-like particles that harbor misfolded proteins and are decorated by protein quality control factors. The constituent proteins of the condensates/aggregates are disentangled by protein disaggregation systems mainly based on Hsp70 and AAA ATPase Hsp100 chaperones prior to their handover to refolding and degradation systems. Here, we discuss the functional roles that condensate formation/aggregation and disaggregation play in protein quality control to maintain proteostasis and why it matters for understanding health and disease.
Collapse
Affiliation(s)
- Verena Kohler
- Institute of Molecular Biosciences, University of Graz, Graz, Austria
| | - Claes Andréasson
- Department of Molecular Biosciences, Stockholm University, Stockholm, Sweden
| |
Collapse
|
16
|
Regulation of germline proteostasis by HSF1 and insulin/IGF-1 signaling. Biochem Soc Trans 2023; 51:501-512. [PMID: 36892215 DOI: 10.1042/bst20220616] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2023] [Revised: 02/14/2023] [Accepted: 02/16/2023] [Indexed: 03/10/2023]
Abstract
Protein homeostasis (proteostasis) is essential for cellular function and organismal health and requires the concerted actions of protein synthesis, folding, transport, and turnover. In sexually reproducing organisms, the immortal germline lineage passes genetic information across generations. Accumulating evidence indicates the importance of proteome integrity for germ cells as genome stability. As gametogenesis involves very active protein synthesis and is highly energy-demanding, it has unique requirements for proteostasis regulation and is sensitive to stress and nutrient availability. The heat shock factor 1 (HSF1), a key transcriptional regulator of cellular response to cytosolic and nuclear protein misfolding has evolutionarily conserved roles in germline development. Similarly, insulin/insulin-like growth factor-1 (IGF-1) signaling, a major nutrient-sensing pathway, impacts many aspects of gametogenesis. Here, we focus on HSF1 and IIS to review insights into their roles in germline proteostasis and discuss the implications on gamete quality control during stress and aging.
Collapse
|
17
|
Lei T, Xiao Z, Bi W, Cai S, Yang Y, Du H. Targeting small heat shock proteins to degrade aggregates as a potential strategy in neurodegenerative diseases. Ageing Res Rev 2022; 82:101769. [PMID: 36283618 DOI: 10.1016/j.arr.2022.101769] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2022] [Revised: 10/18/2022] [Accepted: 10/21/2022] [Indexed: 01/31/2023]
Abstract
Neurodegenerative diseases (NDs) are aging-related diseases that involve the death of neurons in the brain. Dysregulation of protein homeostasis leads to the production of toxic proteins or the formation of aggregates, which is the pathological basis of NDs. Small heat shock proteins (HSPB) is involved in the establishment of a protein quality control (PQC) system to maintain cellular homeostasis. HSPB can be secreted into the extracellular space and delivered by various routes, especially extracellular vehicles (EVs). HSPB plays an important role in influencing the aggregation phase of toxic proteins involved in heat shock transcription factor (HSF) regulation, oxidative stress, autophagy and apoptosis pathways. HSPB conferred neuroprotective effects by resisting toxic protein aggregation, reducing autophagy and reducing neuronal apoptosis. The HSPB treatment strategies, including targeted PQC system therapy and delivery of EVs-HSPB, can improve disease manifestations for NDs. This review aims to provide a comprehensive insight into the impact of HSPB in NDs and the feasibility of new technology to enhance HSPB expression and EVs-HSPB delivery for neurodegenerative disease.
Collapse
Affiliation(s)
- Tong Lei
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Zhuangzhuang Xiao
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Wangyu Bi
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Shanglin Cai
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Yanjie Yang
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China
| | - Hongwu Du
- School of Chemistry and Biological Engineering, University of Science and Technology Beijing, Beijing 100083, China; Daxing Research Institute, University of Science and Technology Beijing, Beijing 100083, China.
| |
Collapse
|
18
|
Ye N, Wang B, Feng W, Tang D, Zeng Z. PRRS virus receptors and an alternative pathway for viral invasion. Virus Res 2022; 320:198885. [PMID: 35948131 DOI: 10.1016/j.virusres.2022.198885] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2022] [Revised: 08/05/2022] [Accepted: 08/06/2022] [Indexed: 11/25/2022]
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV) has a highly restricted cell tropism, which is closely related to the specific receptors associated with PRRSV infection. At least nine cellular molecules have been identified as putative receptors for PRRSV, including CD163, a cysteine-rich scavenger receptor. With the participation of the CD163 receptor and other cofactors, PRRSV invades cells via low pH-dependent clathrin-mediated endocytosis. In addition, PRRSV utilizes viral apoptotic mimicry to infect cells though macropinocytosis as an alternative pathway. In this review, we discuss recent advances in the studies on receptors and pathways that play an important role in PRRSV invasion, and simultaneously explore the use of specific antibodies, small molecules, and blockers targeting receptor-ligand interactions, as a potential strategy for controlling PRRSV infection. Novel antiviral strategies against PRRSV could be developed by identifying the interaction between receptors and ligands.
Collapse
Affiliation(s)
- Ni Ye
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Bin Wang
- College of Animal Science, Guizhou University, Guiyang 550025, China.
| | - Wei Feng
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Deyuan Tang
- College of Animal Science, Guizhou University, Guiyang 550025, China
| | - Zhiyong Zeng
- College of Animal Science, Guizhou University, Guiyang 550025, China
| |
Collapse
|
19
|
Krause GJ, Diaz A, Jafari M, Khawaja RR, Agullo‐Pascual E, Santiago‐Fernández O, Richards AL, Chen K, Dmitriev P, Sun Y, See SK, Abdelmohsen K, Mazan‐Mamczarz K, Krogan NJ, Gorospe M, Swaney DL, Sidoli S, Bravo‐Cordero JJ, Kampmann M, Cuervo AM. Reduced endosomal microautophagy activity in aging associates with enhanced exocyst-mediated protein secretion. Aging Cell 2022; 21:e13713. [PMID: 36116133 PMCID: PMC9577956 DOI: 10.1111/acel.13713] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2022] [Revised: 08/23/2022] [Accepted: 08/24/2022] [Indexed: 01/31/2023] Open
Abstract
Autophagy is essential for protein quality control and regulation of the functional proteome. Failure of autophagy pathways with age contributes to loss of proteostasis in aged organisms and accelerates the progression of age-related diseases. In this work, we show that activity of endosomal microautophagy (eMI), a selective type of autophagy occurring in late endosomes, declines with age and identify the sub-proteome affected by this loss of function. Proteomics of late endosomes from old mice revealed an aberrant glycation signature for Hsc70, the chaperone responsible for substrate targeting to eMI. Age-related Hsc70 glycation reduces its stability in late endosomes by favoring its organization into high molecular weight protein complexes and promoting its internalization/degradation inside late endosomes. Reduction of eMI with age associates with an increase in protein secretion, as late endosomes can release protein-loaded exosomes upon plasma membrane fusion. Our search for molecular mediators of the eMI/secretion switch identified the exocyst-RalA complex, known for its role in exocytosis, as a novel physiological eMI inhibitor that interacts with Hsc70 and acts directly at the late endosome membrane. This inhibitory function along with the higher exocyst-RalA complex levels detected in late endosomes from old mice could explain, at least in part, reduced eMI activity with age. Interaction of Hsc70 with components of the exocyst-RalA complex places this chaperone in the switch from eMI to secretion. Reduced intracellular degradation in favor of extracellular release of undegraded material with age may be relevant to the spreading of proteotoxicity associated with aging and progression of proteinopathies.
Collapse
Affiliation(s)
- Gregory J. Krause
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineBronxNew YorkUSA
- Institute for Aging Studies, Albert Einstein College of MedicineBronxNew YorkUSA
| | - Antonio Diaz
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineBronxNew YorkUSA
- Institute for Aging Studies, Albert Einstein College of MedicineBronxNew YorkUSA
| | - Maryam Jafari
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineBronxNew YorkUSA
- Institute for Aging Studies, Albert Einstein College of MedicineBronxNew YorkUSA
| | - Rabia R. Khawaja
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineBronxNew YorkUSA
- Institute for Aging Studies, Albert Einstein College of MedicineBronxNew YorkUSA
| | - Esperanza Agullo‐Pascual
- Microscopy and Advanced Bioimaging Core, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Olaya Santiago‐Fernández
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineBronxNew YorkUSA
- Institute for Aging Studies, Albert Einstein College of MedicineBronxNew YorkUSA
| | - Alicia L. Richards
- Department of Cellular Molecular PharmacologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- The J. David Gladstone InstitutesSan FranciscoCaliforniaUSA
- Quantitative Biosciences Institute (QBI), University of California San FranciscoSan FranciscoCaliforniaUSA
| | - Kuei‐Ho Chen
- Department of Cellular Molecular PharmacologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- The J. David Gladstone InstitutesSan FranciscoCaliforniaUSA
- Quantitative Biosciences Institute (QBI), University of California San FranciscoSan FranciscoCaliforniaUSA
| | - Phillip Dmitriev
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineBronxNew YorkUSA
- Institute for Aging Studies, Albert Einstein College of MedicineBronxNew YorkUSA
| | - Yan Sun
- Department of BiochemistryAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Stephanie K. See
- Department of Biochemistry and BiophysicsInstitute for Neurodegenerative Diseases, University of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Kotb Abdelmohsen
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research ProgramNational Institutes of HealthBaltimoreMarylandUSA
| | - Krystyna Mazan‐Mamczarz
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research ProgramNational Institutes of HealthBaltimoreMarylandUSA
| | - Nevan J. Krogan
- Department of Cellular Molecular PharmacologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- The J. David Gladstone InstitutesSan FranciscoCaliforniaUSA
- Quantitative Biosciences Institute (QBI), University of California San FranciscoSan FranciscoCaliforniaUSA
| | - Myriam Gorospe
- Laboratory of Genetics and Genomics, National Institute on Aging Intramural Research ProgramNational Institutes of HealthBaltimoreMarylandUSA
| | - Danielle L. Swaney
- Department of Cellular Molecular PharmacologyUniversity of California San FranciscoSan FranciscoCaliforniaUSA
- The J. David Gladstone InstitutesSan FranciscoCaliforniaUSA
- Quantitative Biosciences Institute (QBI), University of California San FranciscoSan FranciscoCaliforniaUSA
| | - Simone Sidoli
- Department of BiochemistryAlbert Einstein College of MedicineBronxNew YorkUSA
| | - Jose Javier Bravo‐Cordero
- Department of Medicine, Division of Hematology and Medical OncologyIcahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
- The Tisch Cancer Institute, Icahn School of Medicine at Mount SinaiNew YorkNew YorkUSA
| | - Martin Kampmann
- Department of Biochemistry and BiophysicsInstitute for Neurodegenerative Diseases, University of California, San FranciscoSan FranciscoCaliforniaUSA
| | - Ana Maria Cuervo
- Department of Developmental and Molecular BiologyAlbert Einstein College of MedicineBronxNew YorkUSA
- Institute for Aging Studies, Albert Einstein College of MedicineBronxNew YorkUSA
| |
Collapse
|
20
|
Calabrese G, Molzahn C, Mayor T. Protein interaction networks in neurodegenerative diseases: from physiological function to aggregation. J Biol Chem 2022; 298:102062. [PMID: 35623389 PMCID: PMC9234719 DOI: 10.1016/j.jbc.2022.102062] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2022] [Revised: 04/26/2022] [Accepted: 05/18/2022] [Indexed: 11/25/2022] Open
Abstract
The accumulation of protein inclusions is linked to many neurodegenerative diseases that typically develop in older individuals, due to a combination of genetic and environmental factors. In rare familial neurodegenerative disorders, genes encoding for aggregation-prone proteins are often mutated. While the underlying mechanism leading to these diseases still remains to be fully elucidated, efforts in the past 20 years revealed a vast network of protein–protein interactions that play a major role in regulating the aggregation of key proteins associated with neurodegeneration. Misfolded proteins that can oligomerize and form insoluble aggregates associate with molecular chaperones and other elements of the proteolytic machineries that are the frontline workers attempting to protect the cells by promoting clearance and preventing aggregation. Proteins that are normally bound to aggregation-prone proteins can become sequestered and mislocalized in protein inclusions, leading to their loss of function. In contrast, mutations, posttranslational modifications, or misfolding of aggregation-prone proteins can lead to gain of function by inducing novel or altered protein interactions, which in turn can impact numerous essential cellular processes and organelles, such as vesicle trafficking and the mitochondria. This review examines our current knowledge of protein–protein interactions involving several key aggregation-prone proteins that are associated with Alzheimer’s disease, Parkinson’s disease, Huntington’s disease, or amyotrophic lateral sclerosis. We aim to provide an overview of the protein interaction networks that play a central role in driving or mitigating inclusion formation, while highlighting some of the key proteomic studies that helped to uncover the extent of these networks.
Collapse
Affiliation(s)
- Gaetano Calabrese
- Michael Smith Laboratories, University of British Columbia, V6T 1Z4 Vancouver BC, Canada.
| | - Cristen Molzahn
- Michael Smith Laboratories, University of British Columbia, V6T 1Z4 Vancouver BC, Canada
| | - Thibault Mayor
- Michael Smith Laboratories, University of British Columbia, V6T 1Z4 Vancouver BC, Canada.
| |
Collapse
|
21
|
Heat Shock Protein Member 8 (HSPA8) Is Involved in Porcine Reproductive and Respiratory Syndrome Virus Attachment and Internalization. Microbiol Spectr 2022; 10:e0186021. [PMID: 35138165 PMCID: PMC8826899 DOI: 10.1128/spectrum.01860-21] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Porcine reproductive and respiratory syndrome virus (PRRSV), a porcine arterivirus, causes severe financial losses to global swine industry. Despite much research, the molecular mechanisms of PRRSV infection remains to be fully elucidated. In the current study, we uncovered the involvement of heat shock protein member 8 (HSPA8) in PRRSV attachment and internalization during infection for the first time. In detail, HSPA8 was identified to interact with PRRSV glycoprotein 4 (GP4), a major determinant for viral cellular tropism, dependent on its carboxy-terminal peptide-binding (PB) domain. Chemical inhibitors and specific small interference RNAs (siRNAs) targeting HSPA8 significantly suppressed PRRSV infection as indicated by decreased viral RNA abundance, infectivity, and titers. Especially, PRRSV attachment was inhibited by interference of its binding to HSPA8 with mouse anti-HSPA8 polyclonal antibodies (pAbs) and recombinant soluble HSPA8 protein. HSPA8 was further shown to participate in PRRSV internalization through clathrin-dependent endocytosis (CME). Collectively, these results demonstrate that HSPA8 is important for PRRSV attachment and internalization, which is a potential target to prevent and control the viral infection. IMPORTANCE PRRSV has caused huge economic losses to the pork industry around the world. Currently, safe and effective strategies are still urgently required to prevent and control PRRSV infection. As the first steps, PRRSV attachment and internalization are initiated by interactions between viral envelope proteins and host cell receptors/factors, which are not fully understood yet. Here, we identified the interaction between PRRSV GP4 and HSPA8, and demonstrated that HSPA8 was involved in PRRSV attachment and internalization. This work deepens our understanding of the molecular mechanisms involved in PRRSV infection, and provides novel insights for the development of antiviral drugs and vaccines against the virus.
Collapse
|
22
|
Prichard KL, O'Brien NS, Murcia SR, Baker JR, McCluskey A. Role of Clathrin and Dynamin in Clathrin Mediated Endocytosis/Synaptic Vesicle Recycling and Implications in Neurological Diseases. Front Cell Neurosci 2022; 15:754110. [PMID: 35115907 PMCID: PMC8805674 DOI: 10.3389/fncel.2021.754110] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Accepted: 12/10/2021] [Indexed: 12/17/2022] Open
Abstract
Endocytosis is a process essential to the health and well-being of cell. It is required for the internalisation and sorting of “cargo”—the macromolecules, proteins, receptors and lipids of cell signalling. Clathrin mediated endocytosis (CME) is one of the key processes required for cellular well-being and signalling pathway activation. CME is key role to the recycling of synaptic vesicles [synaptic vesicle recycling (SVR)] in the brain, it is pivotal to signalling across synapses enabling intracellular communication in the sensory and nervous systems. In this review we provide an overview of the general process of CME with a particular focus on two key proteins: clathrin and dynamin that have a central role to play in ensuing successful completion of CME. We examine these two proteins as they are the two endocytotic proteins for which small molecule inhibitors, often of known mechanism of action, have been identified. Inhibition of CME offers the potential to develop therapeutic interventions into conditions involving defects in CME. This review will discuss the roles and the current scope of inhibitors of clathrin and dynamin, providing an insight into how further developments could affect neurological disease treatments.
Collapse
|
23
|
Unzipping the Secrets of Amyloid Disassembly by the Human Disaggregase. Cells 2021; 10:cells10102745. [PMID: 34685723 PMCID: PMC8534776 DOI: 10.3390/cells10102745] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2021] [Revised: 10/08/2021] [Accepted: 10/08/2021] [Indexed: 01/08/2023] Open
Abstract
Neurodegenerative diseases (NDs) are increasingly positioned as leading causes of global deaths. The accelerated aging of the population and its strong relationship with neurodegeneration forecast these pathologies as a huge global health problem in the upcoming years. In this scenario, there is an urgent need for understanding the basic molecular mechanisms associated with such diseases. A major molecular hallmark of most NDs is the accumulation of insoluble and toxic protein aggregates, known as amyloids, in extracellular or intracellular deposits. Here, we review the current knowledge on how molecular chaperones, and more specifically a ternary protein complex referred to as the human disaggregase, deals with amyloids. This machinery, composed of the constitutive Hsp70 (Hsc70), the class B J-protein DnaJB1 and the nucleotide exchange factor Apg2 (Hsp110), disassembles amyloids of α-synuclein implicated in Parkinson’s disease as well as of other disease-associated proteins such as tau and huntingtin. We highlight recent studies that have led to the dissection of the mechanism used by this chaperone system to perform its disaggregase activity. We also discuss whether this chaperone-mediated disassembly mechanism could be used to solubilize other amyloidogenic substrates. Finally, we evaluate the implications of the chaperone system in amyloid clearance and associated toxicity, which could be critical for the development of new therapies.
Collapse
|
24
|
Nachman E, Verstreken P. Synaptic proteostasis in Parkinson's disease. Curr Opin Neurobiol 2021; 72:72-79. [PMID: 34653835 DOI: 10.1016/j.conb.2021.09.001] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2021] [Accepted: 09/07/2021] [Indexed: 12/20/2022]
Abstract
There are over 7 million people worldwide suffering from Parkinson's disease, and this number will double in the next decade. Causative mutations and risk variants in >20 genes that predominantly act at synapses have been linked to Parkinson's disease. Synaptic defects precede neuronal death. However, we are only now beginning to understand which molecular mechanisms contribute to this synaptic dysfunction. In this review, we discuss recent data demonstrating that Parkinson proteins act centrally to various protein quality control pathways at the synapse, and we argue that disturbed synaptic proteostasis is an early driver of neurodegeneration in Parkinson's disease.
Collapse
Affiliation(s)
- Eliana Nachman
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Herestraat 49, Box 602, 3000 Leuven, Belgium
| | - Patrik Verstreken
- VIB Center for Brain & Disease Research, 3000 Leuven, Belgium; KU Leuven, Department of Neurosciences, Leuven Brain Institute, Mission Lucidity, Herestraat 49, Box 602, 3000 Leuven, Belgium.
| |
Collapse
|
25
|
Silva NSM, Rodrigues LFDC, Dores-Silva PR, Montanari CA, Ramos CHI, Barbosa LRS, Borges JC. Structural, thermodynamic and functional studies of human 71 kDa heat shock cognate protein (HSPA8/hHsc70). BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2021; 1869:140719. [PMID: 34571256 DOI: 10.1016/j.bbapap.2021.140719] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/01/2021] [Revised: 08/29/2021] [Accepted: 09/21/2021] [Indexed: 01/14/2023]
Abstract
Human 71 kDa heat shock cognate protein (HSPA8, also known as Hsc70, Hsp70-8, Hsc71, Hsp71 or Hsp73) is a constitutively expressed chaperone that is critical for cell proteostasis. In the cytosol, HSPA8 plays a pivotal role in folding and refolding, facilitates protein trafficking across membranes and targets proteins for degradation, among other functions. Here, we report an in solution study of recombinant HSPA8 (rHSPA8) using a variety of biophysical and biochemical approaches. rHSPA8 shares several structural and functional similarities with others human Hsp70s. It has two domains with different stabilities and interacts with adenosine nucleotides with dissociation constants in the low micromolar range, which were higher in the presence of Mg2+. rHSPA8 showed lower ATPase activity than its homolog HSPA5/hGrp78/hBiP, but it was 4-fold greater than that of recombinant HSPA1A/hHsp70-1A, with which it is 86% identical. Small angle X-ray scattering indicated that rHSPA8 behaved as an elongated monomeric protein in solution with dimensions similar to those observed for HSPA1A. In addition, rHSPA8 showed structural flexibility between its compacted and extended conformations. The data also indicated that HSPA8 has capacity in preventing the aggregation of model client proteins. The present study expands the understanding of the structure and activity of this chaperone and aligns with the idea that human homologous Hsp70s have divergent functions.
Collapse
Affiliation(s)
| | | | - Paulo Roberto Dores-Silva
- São Carlos Institute of Chemistry, University of São Paulo, São Carlos, SP, Brazil; Division of Trauma, Critical Care, Burns and Acute Care Surgery, Department of Surgery School of Medicine, University of California San Diego, La Jolla, CA 92093, USA
| | | | | | - Leandro Ramos Souza Barbosa
- Institute of Physics, University of São Paulo, São Paulo, SP, Brazil; Brazilian Synchrotron Light Laboratory (LNLS), Brazilian Center for Research in Energy and Materials (CNPEM), Campinas, Brazil
| | - Júlio César Borges
- São Carlos Institute of Chemistry, University of São Paulo, São Carlos, SP, Brazil.
| |
Collapse
|
26
|
Robbins M, Clayton E, Kaminski Schierle GS. Synaptic tau: A pathological or physiological phenomenon? Acta Neuropathol Commun 2021; 9:149. [PMID: 34503576 PMCID: PMC8428049 DOI: 10.1186/s40478-021-01246-y] [Citation(s) in RCA: 54] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Accepted: 08/12/2021] [Indexed: 12/17/2022] Open
Abstract
In this review, we discuss the synaptic aspects of Tau pathology occurring during Alzheimer's disease (AD) and how this may relate to memory impairment, a major hallmark of AD. Whilst the clinical diagnosis of AD patients is a loss of working memory and long-term declarative memory, the histological diagnosis is the presence of neurofibrillary tangles of hyperphosphorylated Tau and Amyloid-beta plaques. Tau pathology spreads through synaptically connected neurons to impair synaptic function preceding the formation of neurofibrillary tangles, synaptic loss, axonal retraction and cell death. Alongside synaptic pathology, recent data suggest that Tau has physiological roles in the pre- or post- synaptic compartments. Thus, we have seen a shift in the research focus from Tau as a microtubule-stabilising protein in axons, to Tau as a synaptic protein with roles in accelerating spine formation, dendritic elongation, and in synaptic plasticity coordinating memory pathways. We collate here the myriad of emerging interactions and physiological roles of synaptic Tau, and discuss the current evidence that synaptic Tau contributes to pathology in AD.
Collapse
|
27
|
Powers ET, Gierasch LM. The Proteome Folding Problem and Cellular Proteostasis. J Mol Biol 2021; 433:167197. [PMID: 34391802 DOI: 10.1016/j.jmb.2021.167197] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 08/04/2021] [Accepted: 08/04/2021] [Indexed: 12/16/2022]
Abstract
Stunning advances have been achieved in addressing the protein folding problem, providing deeper understanding of the mechanisms by which proteins navigate energy landscapes to reach their native states and enabling powerful algorithms to connect sequence to structure. However, the realities of the in vivo protein folding problem remain a challenge to reckon with. Here, we discuss the concept of the "proteome folding problem"-the problem of how organisms build and maintain a functional proteome-by admitting that folding energy landscapes are characterized by many misfolded states and that cells must deploy a network of chaperones and degradation enzymes to minimize deleterious impacts of these off-pathway species. The resulting proteostasis network is an inextricable part of in vivo protein folding and must be understood in detail if we are to solve the proteome folding problem. We discuss how the development of computational models for the proteostasis network's actions and the relationship to the biophysical properties of the proteome has begun to offer new insights and capabilities.
Collapse
Affiliation(s)
- Evan T Powers
- Department of Chemistry, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | - Lila M Gierasch
- Departments of Biochemistry & Molecular Biology and Chemistry, University of Massachusetts-Amherst, Amherst, MA 01003, USA.
| |
Collapse
|
28
|
AgDD System: A Chemical Controllable Protein Aggregates in Cells. Methods Mol Biol 2021. [PMID: 34228296 DOI: 10.1007/978-1-0716-1441-9_16] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register]
Abstract
There are increasing evidence and growing interest in the relationship between protein aggregates/phase separation and various human diseases, especially neurodegenerative diseases. However, we do not entirely comprehend how aggregates generate or the clearance network of chaperones, proteasomes, ubiquitin ligases, and other factors interact with aggregates. Here, we describe chemically controllable systems compose with a genetically engineered cell and a small drug that enables us to rapidly induce protein aggregates' formation by withdrawing the small molecule. This trigger does not activate global stress responses induced by stimuli, such as proteasome inhibitors or heat shock. This method can produce aggregates in a specific compartment and diverse experimental systems, including live animals.
Collapse
|
29
|
Ruano D. Proteostasis Dysfunction in Aged Mammalian Cells. The Stressful Role of Inflammation. Front Mol Biosci 2021; 8:658742. [PMID: 34222330 PMCID: PMC8245766 DOI: 10.3389/fmolb.2021.658742] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2021] [Accepted: 05/28/2021] [Indexed: 12/15/2022] Open
Abstract
Aging is a biological and multifactorial process characterized by a progressive and irreversible deterioration of the physiological functions leading to a progressive increase in morbidity. In the next decades, the world population is expected to reach ten billion, and globally, elderly people over 80 are projected to triple in 2050. Consequently, it is also expected an increase in the incidence of age-related pathologies such as cancer, diabetes, or neurodegenerative disorders. Disturbance of cellular protein homeostasis (proteostasis) is a hallmark of normal aging that increases cell vulnerability and might be involved in the etiology of several age-related diseases. This review will focus on the molecular alterations occurring during normal aging in the most relevant protein quality control systems such as molecular chaperones, the UPS, and the ALS. Also, alterations in their functional cooperation will be analyzed. Finally, the role of inflammation, as a synergistic negative factor of the protein quality control systems during normal aging, will also be addressed. A better comprehension of the age-dependent modifications affecting the cellular proteostasis, as well as the knowledge of the mechanisms underlying these alterations, might be very helpful to identify relevant risk factors that could be responsible for or contribute to cell deterioration, a fundamental question still pending in biomedicine.
Collapse
Affiliation(s)
- Diego Ruano
- Instituto de Biomedicina de Sevilla (IBiS), Hospital Universitario Virgen del Rocío/Consejo Superior de Investigaciones Científicas/Universidad de Sevilla, Sevilla, Spain.,Departamento de Bioquímica y Biología Molecular, Facultad de Farmacia, Universidad de Sevilla, Sevilla, Spain
| |
Collapse
|
30
|
Bourdenx M, Martín-Segura A, Scrivo A, Rodriguez-Navarro JA, Kaushik S, Tasset I, Diaz A, Storm NJ, Xin Q, Juste YR, Stevenson E, Luengo E, Clement CC, Choi SJ, Krogan NJ, Mosharov EV, Santambrogio L, Grueninger F, Collin L, Swaney DL, Sulzer D, Gavathiotis E, Cuervo AM. Chaperone-mediated autophagy prevents collapse of the neuronal metastable proteome. Cell 2021; 184:2696-2714.e25. [PMID: 33891876 DOI: 10.1016/j.cell.2021.03.048] [Citation(s) in RCA: 209] [Impact Index Per Article: 52.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2020] [Revised: 01/03/2021] [Accepted: 03/23/2021] [Indexed: 12/18/2022]
Abstract
Components of the proteostasis network malfunction in aging, and reduced protein quality control in neurons has been proposed to promote neurodegeneration. Here, we investigate the role of chaperone-mediated autophagy (CMA), a selective autophagy shown to degrade neurodegeneration-related proteins, in neuronal proteostasis. Using mouse models with systemic and neuronal-specific CMA blockage, we demonstrate that loss of neuronal CMA leads to altered neuronal function, selective changes in the neuronal metastable proteome, and proteotoxicity, all reminiscent of brain aging. Imposing CMA loss on a mouse model of Alzheimer's disease (AD) has synergistic negative effects on the proteome at risk of aggregation, thus increasing neuronal disease vulnerability and accelerating disease progression. Conversely, chemical enhancement of CMA ameliorates pathology in two different AD experimental mouse models. We conclude that functional CMA is essential for neuronal proteostasis through the maintenance of a subset of the proteome with a higher risk of misfolding than the general proteome.
Collapse
Affiliation(s)
- Mathieu Bourdenx
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Adrián Martín-Segura
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Aurora Scrivo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Jose A Rodriguez-Navarro
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Susmita Kaushik
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Inmaculada Tasset
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Antonio Diaz
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Nadia J Storm
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Qisheng Xin
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA
| | - Yves R Juste
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Erica Stevenson
- Department of Cellular Molecular Pharmacology, School of Medicine and California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158, USA; David Gladstone Institutes, San Francisco, CA 94158, USA
| | - Enrique Luengo
- Department of Pharmacology, School of Medicine, Instituto Teófilo Hernando for Drug Discovery, Universidad Autonoma de Madrid, Madrid 28049, Spain
| | - Cristina C Clement
- Department of Radiation Oncology, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Se Joon Choi
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY 10461, USA
| | - Nevan J Krogan
- Department of Cellular Molecular Pharmacology, School of Medicine and California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158, USA; David Gladstone Institutes, San Francisco, CA 94158, USA
| | - Eugene V Mosharov
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY 10461, USA
| | - Laura Santambrogio
- Department of Radiation Oncology, Englander Institute for Precision Medicine, Weill Cornell Medicine, New York, NY 10021, USA
| | - Fiona Grueninger
- Roche Pharma Research and Early Development (pRED), Neuro-Immunology, Roche Innovation Center Basel, CH-4070, Switzerland
| | - Ludovic Collin
- Roche Pharma Research and Early Development (pRED), Neuro-Immunology, Roche Innovation Center Basel, CH-4070, Switzerland
| | - Danielle L Swaney
- Department of Cellular Molecular Pharmacology, School of Medicine and California Institute for Quantitative Biosciences, University of California, San Francisco, San Francisco, CA 94158, USA; David Gladstone Institutes, San Francisco, CA 94158, USA
| | - David Sulzer
- Department of Psychiatry, Columbia University Medical Center, New York State Psychiatric Institute, New York, NY 10461, USA; Departments of Neurology and Pharmacology, Columbia University Medical Center, New York, NY 10032, USA
| | - Evripidis Gavathiotis
- Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA; Department of Biochemistry, Albert Einstein College of Medicine, Bronx, New York, NY 10461, USA; Department of Medicine, Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, Bronx, NY 10461, USA; Institute for Aging Studies of the Department of Medicine of the Albert Einstein College of Medicine, Bronx, NY 10461, USA.
| |
Collapse
|
31
|
Plagens RN, Mossiah I, Kim Guisbert KS, Guisbert E. Chronic temperature stress inhibits reproduction and disrupts endocytosis via chaperone titration in Caenorhabditis elegans. BMC Biol 2021; 19:75. [PMID: 33858388 PMCID: PMC8051109 DOI: 10.1186/s12915-021-01008-1] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2021] [Accepted: 03/19/2021] [Indexed: 11/24/2022] Open
Abstract
Background Temperature influences biology at all levels, from altering rates of biochemical reactions to determining sustainability of entire ecosystems. Although extended exposure to elevated temperatures influences organismal phenotypes important for human health, agriculture, and ecology, the molecular mechanisms that drive these responses remain largely unexplored. Prolonged, mild temperature stress (48 h at 28 °C) has been shown to inhibit reproduction in Caenorhabditis elegans without significantly impacting motility or viability. Results Analysis of molecular responses to chronic stress using RNA-seq uncovers dramatic effects on the transcriptome that are fundamentally distinct from the well-characterized, acute heat shock response (HSR). While a large portion of the genome is differentially expressed ≥ 4-fold after 48 h at 28 °C, the only major class of oogenesis-associated genes affected is the vitellogenin gene family that encodes for yolk proteins (YPs). Whereas YP mRNAs decrease, the proteins accumulate and mislocalize in the pseudocoelomic space as early as 6 h, well before reproduction declines. A trafficking defect in a second, unrelated fluorescent reporter and a decrease in pre-synaptic neuronal signaling indicate that the YP mislocalization is caused by a generalized defect in endocytosis. Molecular chaperones are involved in both endocytosis and refolding damaged proteins. Decreasing levels of the major HSP70 chaperone, HSP-1, causes similar YP trafficking defects in the absence of stress. Conversely, increasing chaperone levels through overexpression of the transcription factor HSF-1 rescues YP trafficking and restores neuronal signaling. Conclusions These data implicate chaperone titration during chronic stress as a molecular mechanism contributing to endocytic defects that influence multiple aspects of organismal physiology. Notably, HSF-1 overexpression improves recovery of viable offspring after exposure to stress. These findings provide important molecular insights into understanding organismal responses to temperature stress as well as phenotypes associated with chronic protein misfolding. Supplementary Information The online version contains supplementary material available at 10.1186/s12915-021-01008-1.
Collapse
Affiliation(s)
- Rosemary N Plagens
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, USA
| | - Isiah Mossiah
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, USA
| | - Karen S Kim Guisbert
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, USA
| | - Eric Guisbert
- Department of Biomedical and Chemical Engineering and Sciences, Florida Institute of Technology, Melbourne, FL, USA.
| |
Collapse
|
32
|
Shemesh N, Jubran J, Dror S, Simonovsky E, Basha O, Argov C, Hekselman I, Abu-Qarn M, Vinogradov E, Mauer O, Tiago T, Carra S, Ben-Zvi A, Yeger-Lotem E. The landscape of molecular chaperones across human tissues reveals a layered architecture of core and variable chaperones. Nat Commun 2021; 12:2180. [PMID: 33846299 PMCID: PMC8042005 DOI: 10.1038/s41467-021-22369-9] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 02/23/2021] [Indexed: 12/13/2022] Open
Abstract
The sensitivity of the protein-folding environment to chaperone disruption can be highly tissue-specific. Yet, the organization of the chaperone system across physiological human tissues has received little attention. Through computational analyses of large-scale tissue transcriptomes, we unveil that the chaperone system is composed of core elements that are uniformly expressed across tissues, and variable elements that are differentially expressed to fit with tissue-specific requirements. We demonstrate via a proteomic analysis that the muscle-specific signature is functional and conserved. Core chaperones are significantly more abundant across tissues and more important for cell survival than variable chaperones. Together with variable chaperones, they form tissue-specific functional networks. Analysis of human organ development and aging brain transcriptomes reveals that these functional networks are established in development and decline with age. In this work, we expand the known functional organization of de novo versus stress-inducible eukaryotic chaperones into a layered core-variable architecture in multi-cellular organisms.
Collapse
Affiliation(s)
- Netta Shemesh
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel.,Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Juman Jubran
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Shiran Dror
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Eyal Simonovsky
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Omer Basha
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Chanan Argov
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Idan Hekselman
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Mehtap Abu-Qarn
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Ekaterina Vinogradov
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Omry Mauer
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel
| | - Tatiana Tiago
- Centre for Neuroscience and Nanotechnology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Serena Carra
- Centre for Neuroscience and Nanotechnology, Department of Biomedical, Metabolic and Neural Sciences, University of Modena and Reggio Emilia, Modena, Italy
| | - Anat Ben-Zvi
- Department of Life Sciences, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| | - Esti Yeger-Lotem
- Department of Clinical Biochemistry and Pharmacology and the National Institute for Biotechnology in the Negev, Ben-Gurion University of the Negev, Beer Sheva, Israel.
| |
Collapse
|
33
|
Medina L, González-Lizárraga F, Dominguez-Meijide A, Ploper D, Parrales V, Sequeira S, Cima-Omori MS, Zweckstetter M, Del Bel E, Michel PP, Outeiro TF, Raisman-Vozari R, Chehín R, Socias SB. Doxycycline Interferes With Tau Aggregation and Reduces Its Neuronal Toxicity. Front Aging Neurosci 2021; 13:635760. [PMID: 33828477 PMCID: PMC8020845 DOI: 10.3389/fnagi.2021.635760] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2020] [Accepted: 02/22/2021] [Indexed: 01/15/2023] Open
Abstract
Tauopathies are neurodegenerative disorders with increasing incidence and still without cure. The extensive time required for development and approval of novel therapeutics highlights the need for testing and repurposing known safe molecules. Since doxycycline impacts α-synuclein aggregation and toxicity, herein we tested its effect on tau. We found that doxycycline reduces amyloid aggregation of the 2N4R and K18 isoforms of tau protein in a dose-dependent manner. Furthermore, in a cell free system doxycycline also prevents tau seeding and in cell culture reduces toxicity of tau aggregates. Overall, our results expand the spectrum of action of doxycycline against aggregation-prone proteins, opening novel perspectives for its repurposing as a disease-modifying drug for tauopathies.
Collapse
Affiliation(s)
- Luciana Medina
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (UNT-CONICET-SIPROSA), Tucumán, Argentina
| | - Florencia González-Lizárraga
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (UNT-CONICET-SIPROSA), Tucumán, Argentina
| | - Antonio Dominguez-Meijide
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany.,Laboratory of Neuroanatomy and Experimental Neurology, Department of Morphological Sciences, Center for Research in Molecular Medicine and Chronic Diseases (CIMUS), Instituto de Investigación Sanitaria de Santiago de Compostela (IDIS), University of Santiago de Compostela, Santiago de Compostela, Spain.,Networking Research Center on Neurodegenerative Diseases (CIBERNED), Madrid, Spain
| | - Diego Ploper
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (UNT-CONICET-SIPROSA), Tucumán, Argentina
| | - Valeria Parrales
- Sorbonne Université, Paris Brain Institute -ICM, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Assistance Publique - Hôpitaux de Paris (APHP), Hôpital de la Pitié Salpêtrière, Paris, France
| | - Sabrina Sequeira
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (UNT-CONICET-SIPROSA), Tucumán, Argentina
| | - Maria-Sol Cima-Omori
- German Center for Neurodegenerative Diseases Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany
| | - Markus Zweckstetter
- German Center for Neurodegenerative Diseases Deutsches Zentrum für Neurodegenerative Erkrankungen (DZNE), Göttingen, Germany.,Department for NMR-based Structural Biology, Max Planck Institute for Biophysical Chemistry, Göttingen, Germany
| | - Elaine Del Bel
- Physiology- Dental School of Ribeirão Preto, University of São Paulo, São Paulo, Brazil
| | - Patrick P Michel
- Sorbonne Université, Paris Brain Institute -ICM, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Assistance Publique - Hôpitaux de Paris (APHP), Hôpital de la Pitié Salpêtrière, Paris, France
| | - Tiago Fleming Outeiro
- Department of Experimental Neurodegeneration, Center for Biostructural Imaging of Neurodegeneration, University Medical Center Goettingen, Goettingen, Germany.,Max Planck Institute for Experimental Medicine, Goettingen, Germany.,Translational and Clinical Research Institute, Faculty of Medical Sciences, Newcastle University, Newcastle upon Tyne, United Kingdom
| | - Rita Raisman-Vozari
- Sorbonne Université, Paris Brain Institute -ICM, Institut National de la Santé et de la Recherche Médicale (INSERM), Centre National de la Recherche Scientifique (CNRS), Assistance Publique - Hôpitaux de Paris (APHP), Hôpital de la Pitié Salpêtrière, Paris, France
| | - Rosana Chehín
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (UNT-CONICET-SIPROSA), Tucumán, Argentina
| | - Sergio B Socias
- Instituto de Investigación en Medicina Molecular y Celular Aplicada (IMMCA) (UNT-CONICET-SIPROSA), Tucumán, Argentina
| |
Collapse
|
34
|
Hervás R, Oroz J. Mechanistic Insights into the Role of Molecular Chaperones in Protein Misfolding Diseases: From Molecular Recognition to Amyloid Disassembly. Int J Mol Sci 2020; 21:ijms21239186. [PMID: 33276458 PMCID: PMC7730194 DOI: 10.3390/ijms21239186] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2020] [Revised: 11/29/2020] [Accepted: 11/29/2020] [Indexed: 12/14/2022] Open
Abstract
Age-dependent alterations in the proteostasis network are crucial in the progress of prevalent neurodegenerative diseases, such as Alzheimer’s, Parkinson’s, or amyotrophic lateral sclerosis, which are characterized by the presence of insoluble protein deposits in degenerating neurons. Because molecular chaperones deter misfolded protein aggregation, regulate functional phase separation, and even dissolve noxious aggregates, they are considered major sentinels impeding the molecular processes that lead to cell damage in the course of these diseases. Indeed, members of the chaperome, such as molecular chaperones and co-chaperones, are increasingly recognized as therapeutic targets for the development of treatments against degenerative proteinopathies. Chaperones must recognize diverse toxic clients of different orders (soluble proteins, biomolecular condensates, organized protein aggregates). It is therefore critical to understand the basis of the selective chaperone recognition to discern the mechanisms of action of chaperones in protein conformational diseases. This review aimed to define the selective interplay between chaperones and toxic client proteins and the basis for the protective role of these interactions. The presence and availability of chaperone recognition motifs in soluble proteins and in insoluble aggregates, both functional and pathogenic, are discussed. Finally, the formation of aberrant (pro-toxic) chaperone complexes will also be disclosed.
Collapse
Affiliation(s)
- Rubén Hervás
- Stowers Institute for Medical Research, Kansas City, MO 64110, USA;
| | - Javier Oroz
- Rocasolano Institute for Physical Chemistry, Spanish National Research Council (IQFR-CSIC), Serrano 119, E-28006 Madrid, Spain
- Correspondence: ; Tel.: +34-915619400
| |
Collapse
|
35
|
Ryckman AE, Brockhausen I, Walia JS. Metabolism of Glycosphingolipids and Their Role in the Pathophysiology of Lysosomal Storage Disorders. Int J Mol Sci 2020; 21:E6881. [PMID: 32961778 PMCID: PMC7555265 DOI: 10.3390/ijms21186881] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2020] [Revised: 09/04/2020] [Accepted: 09/12/2020] [Indexed: 12/11/2022] Open
Abstract
Glycosphingolipids (GSLs) are a specialized class of membrane lipids composed of a ceramide backbone and a carbohydrate-rich head group. GSLs populate lipid rafts of the cell membrane of eukaryotic cells, and serve important cellular functions including control of cell-cell signaling, signal transduction and cell recognition. Of the hundreds of unique GSL structures, anionic gangliosides are the most heavily implicated in the pathogenesis of lysosomal storage diseases (LSDs) such as Tay-Sachs and Sandhoff disease. Each LSD is characterized by the accumulation of GSLs in the lysosomes of neurons, which negatively interact with other intracellular molecules to culminate in cell death. In this review, we summarize the biosynthesis and degradation pathways of GSLs, discuss how aberrant GSL metabolism contributes to key features of LSD pathophysiology, draw parallels between LSDs and neurodegenerative proteinopathies such as Alzheimer's and Parkinson's disease and lastly, discuss possible therapies for patients.
Collapse
Affiliation(s)
| | - Inka Brockhausen
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 2V5, Canada;
| | - Jagdeep S. Walia
- Department of Biomedical and Molecular Sciences, Queen’s University, Kingston, ON K7L 2V5, Canada;
| |
Collapse
|
36
|
Walsh MB, Janzen E, Wingrove E, Hosseinibarkooie S, Muela NR, Davidow L, Dimitriadi M, Norabuena EM, Rubin LL, Wirth B, Hart AC. Genetic modifiers ameliorate endocytic and neuromuscular defects in a model of spinal muscular atrophy. BMC Biol 2020; 18:127. [PMID: 32938453 PMCID: PMC7495824 DOI: 10.1186/s12915-020-00845-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Accepted: 08/11/2020] [Indexed: 12/31/2022] Open
Abstract
Background Understanding the genetic modifiers of neurodegenerative diseases can provide insight into the mechanisms underlying these disorders. Here, we examine the relationship between the motor neuron disease spinal muscular atrophy (SMA), which is caused by reduced levels of the survival of motor neuron (SMN) protein, and the actin-bundling protein Plastin 3 (PLS3). Increased PLS3 levels suppress symptoms in a subset of SMA patients and ameliorate defects in SMA disease models, but the functional connection between PLS3 and SMN is poorly understood. Results We provide immunohistochemical and biochemical evidence for large protein complexes localized in vertebrate motor neuron processes that contain PLS3, SMN, and members of the hnRNP F/H family of proteins. Using a Caenorhabditis elegans (C. elegans) SMA model, we determine that overexpression of PLS3 or loss of the C. elegans hnRNP F/H ortholog SYM-2 enhances endocytic function and ameliorates neuromuscular defects caused by decreased SMN-1 levels. Furthermore, either increasing PLS3 or decreasing SYM-2 levels suppresses defects in a C. elegans ALS model. Conclusions We propose that hnRNP F/H act in the same protein complex as PLS3 and SMN and that the function of this complex is critical for endocytic pathways, suggesting that hnRNP F/H proteins could be potential targets for therapy development.
Collapse
Affiliation(s)
- Melissa B Walsh
- Department of Neuroscience, Brown University, 185 Meeting Street, Mailbox GL-N, Providence, RI, 02912, USA
| | - Eva Janzen
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Institute of Genetics, and Center for Rare Disorders, University of Cologne, Cologne, Germany
| | - Emily Wingrove
- Department of Neuroscience, Brown University, 185 Meeting Street, Mailbox GL-N, Providence, RI, 02912, USA
| | - Seyyedmohsen Hosseinibarkooie
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Institute of Genetics, and Center for Rare Disorders, University of Cologne, Cologne, Germany
| | - Natalia Rodriguez Muela
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Lance Davidow
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Maria Dimitriadi
- Department of Biological and Environmental Sciences, University of Hertfordshire, Hertfordshire, UK
| | - Erika M Norabuena
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Lee L Rubin
- Department of Stem Cell & Regenerative Biology, Harvard University, Cambridge, MA, 02138, USA
| | - Brunhilde Wirth
- Institute of Human Genetics, Center for Molecular Medicine Cologne, Institute of Genetics, and Center for Rare Disorders, University of Cologne, Cologne, Germany
| | - Anne C Hart
- Department of Neuroscience, Brown University, 185 Meeting Street, Mailbox GL-N, Providence, RI, 02912, USA.
| |
Collapse
|
37
|
Lualdi M, Alberio T, Fasano M. Proteostasis and Proteotoxicity in the Network Medicine Era. Int J Mol Sci 2020; 21:E6405. [PMID: 32899160 PMCID: PMC7503343 DOI: 10.3390/ijms21176405] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2020] [Revised: 08/28/2020] [Accepted: 09/02/2020] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative proteinopathies are complex diseases that share some pathogenetic processes. One of these is the failure of the proteostasis network (PN), which includes all components involved in the synthesis, folding, and degradation of proteins, thus leading to the aberrant accumulation of toxic protein aggregates in neurons. The single components that belong to the three main modules of the PN are highly interconnected and can be considered as part of a single giant network. Several pharmacological strategies have been proposed to ameliorate neurodegeneration by targeting PN components. Nevertheless, effective disease-modifying therapies are still lacking. In this review article, after a general description of the PN and its failure in proteinopathies, we will focus on the available pharmacological tools to target proteostasis. In this context, we will discuss the main advantages of systems-based pharmacology in contrast to the classical targeted approach, by focusing on network pharmacology as a strategy to innovate rational drug design.
Collapse
Affiliation(s)
| | | | - Mauro Fasano
- Department of Science and High Technology and Center of Bioinformatics, University of Insubria, I-21052 Busto Arsizio, Italy; (M.L.); (T.A.)
| |
Collapse
|
38
|
Niu L, Lou F, Sun Y, Sun L, Cai X, Liu Z, Zhou H, Wang H, Wang Z, Bai J, Yin Q, Zhang J, Chen L, Peng D, Xu Z, Gao Y, Tang S, Fan L, Wang H. A micropeptide encoded by lncRNA MIR155HG suppresses autoimmune inflammation via modulating antigen presentation. SCIENCE ADVANCES 2020; 6:eaaz2059. [PMID: 32671205 PMCID: PMC7314557 DOI: 10.1126/sciadv.aaz2059] [Citation(s) in RCA: 134] [Impact Index Per Article: 26.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/21/2019] [Accepted: 03/06/2020] [Indexed: 05/04/2023]
Abstract
Many annotated long noncoding RNAs (lncRNAs) harbor predicted short open reading frames (sORFs), but the coding capacities of these sORFs and the functions of the resulting micropeptides remain elusive. Here, we report that human lncRNA MIR155HG encodes a 17-amino acid micropeptide, which we termed miPEP155 (P155). MIR155HG is highly expressed by inflamed antigen-presenting cells, leading to the discovery that P155 interacts with the adenosine 5'-triphosphate binding domain of heat shock cognate protein 70 (HSC70), a chaperone required for antigen trafficking and presentation in dendritic cells (DCs). P155 modulates major histocompatibility complex class II-mediated antigen presentation and T cell priming by disrupting the HSC70-HSP90 machinery. Exogenously injected P155 improves two classical mouse models of DC-driven auto inflammation. Collectively, we demonstrate the endogenous existence of a micropeptide encoded by a transcript annotated as "non-protein coding" and characterize a micropeptide as a regulator of antigen presentation and a suppressor of inflammatory diseases.
Collapse
Affiliation(s)
- Liman Niu
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Fangzhou Lou
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Yang Sun
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Libo Sun
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Xiaojie Cai
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Zhaoyuan Liu
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Hong Zhou
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Hong Wang
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Zhikai Wang
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Jing Bai
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Qianqian Yin
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Junxun Zhang
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Linjiao Chen
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Danhong Peng
- Second Clinical Medical College, Guangzhou University of Chinese Medicine, Guangzhou 510006, China
| | - Zhenyao Xu
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Yuanyuan Gao
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Sibei Tang
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Li Fan
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| | - Honglin Wang
- Department of Immunology and Microbiology, Shanghai Institute of Immunology, Key Laboratory of Cell Differentiation and Apoptosis of Chinese Ministry of Education, Institute of Translational Medicine, Shanghai General Hospital, Shanghai Jiao Tong University School of Medicine (SJTU-SM), Shanghai 200025, China
| |
Collapse
|
39
|
Abstract
Ageing is a major risk factor for the development of many diseases, prominently including neurodegenerative disorders such as Alzheimer disease and Parkinson disease. A hallmark of many age-related diseases is the dysfunction in protein homeostasis (proteostasis), leading to the accumulation of protein aggregates. In healthy cells, a complex proteostasis network, comprising molecular chaperones and proteolytic machineries and their regulators, operates to ensure the maintenance of proteostasis. These factors coordinate protein synthesis with polypeptide folding, the conservation of protein conformation and protein degradation. However, sustaining proteome balance is a challenging task in the face of various external and endogenous stresses that accumulate during ageing. These stresses lead to the decline of proteostasis network capacity and proteome integrity. The resulting accumulation of misfolded and aggregated proteins affects, in particular, postmitotic cell types such as neurons, manifesting in disease. Recent analyses of proteome-wide changes that occur during ageing inform strategies to improve proteostasis. The possibilities of pharmacological augmentation of the capacity of proteostasis networks hold great promise for delaying the onset of age-related pathologies associated with proteome deterioration and for extending healthspan.
Collapse
|
40
|
Rosselli-Murai LK, Joseph JG, Lopes-Cendes I, Liu AP, Murai MJ. The Machado-Joseph disease-associated form of ataxin-3 impacts dynamics of clathrin-coated pits. Cell Biol Int 2020; 44:1252-1259. [PMID: 31970864 DOI: 10.1002/cbin.11312] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Accepted: 01/22/2020] [Indexed: 12/14/2022]
Abstract
Expansion above a certain threshold in the polyglutamine (polyQ) tract of ataxin-3 is the main cause of neurodegeneration in Machado-Joseph disease. Ataxin-3 contains an N-terminal catalytic domain, called Josephin domain, and a highly aggregation-prone C-terminal domain containing the polyQ tract. Recent work has shown that protein aggregation inhibits clathrin-mediated endocytosis (CME). However, the effects of polyQ expansion in ataxin-3 on CME have not been investigated. We hypothesize that the expansion of the polyQ tract in ataxin-3 could impact CME. Here, we report that both the wild-type and the expanded ataxin-3 reduce transferrin internalization and expanded ataxin-3 impacts dynamics of clathrin-coated pits (CCPs) by reducing CCP nucleation and increasing short-lived abortive CCPs. Since endocytosis plays a central role in regulating receptor uptake and cargo release, our work highlights a potential mechanism linking protein aggregation to cellular dysregulation.
Collapse
Affiliation(s)
- Luciana K Rosselli-Murai
- Department of Pharmacology, University of Michigan Medical School, 2301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, Michigan, 48109, USA.,Department of Mechanical Engineering, University of Michigan, 2674 GGB, 2350 Hayward, Ann Arbor, Michigan, 48109, USA
| | - Jophin G Joseph
- Department of Mechanical Engineering, University of Michigan, 2674 GGB, 2350 Hayward, Ann Arbor, Michigan, 48109, USA
| | - Iscia Lopes-Cendes
- Department of Medical Genetics, School of Medical Sciences, University of Campinas, R. Tessália Vieira de Camargo, 126, Campinas, São Paulo, 13083-970, Brazil.,The Brazilian Institute of Neuroscience and Neurotechnology, R. Vital Brasil, 251, Campinas, São Paulo, 13083-888, Brazil
| | - Allen P Liu
- Department of Mechanical Engineering, University of Michigan, 2674 GGB, 2350 Hayward, Ann Arbor, Michigan, 48109, USA
| | - Marcelo J Murai
- Department of Pharmacology, University of Michigan Medical School, 2301 MSRB III, 1150 W. Medical Center Dr., Ann Arbor, Michigan, 48109, USA.,Department of Medical Genetics, School of Medical Sciences, University of Campinas, R. Tessália Vieira de Camargo, 126, Campinas, São Paulo, 13083-970, Brazil
| |
Collapse
|
41
|
The Herpes Simplex Virus 1 Immediate Early Protein ICP22 Is a Functional Mimic of a Cellular J Protein. J Virol 2020; 94:JVI.01564-19. [PMID: 31748398 DOI: 10.1128/jvi.01564-19] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2019] [Accepted: 11/14/2019] [Indexed: 12/15/2022] Open
Abstract
Molecular chaperones and cochaperones are the most abundant cellular effectors of protein homeostasis, assisting protein folding and preventing aggregation of misfolded proteins. We have previously shown that herpes simplex virus 1 (HSV-1) infection results in the drastic spatial reorganization of the cellular chaperone Hsc70 into nuclear domains called VICE (Virus Induced Chaperone Enriched) domains and that this recruitment is dependent on the viral immediate early protein ICP22. Here, we present several lines of evidence supporting the notion that ICP22 functions as a virally encoded cochaperone (J-protein/Hsp40) functioning together with its Hsc70 partner to recognize and manage aggregated and misfolded proteins. We show that ICP22 results in (i) nuclear sequestration of nonnative proteins, (ii) reduction of cytoplasmic aggresomes in cells expressing aggregation-prone proteins, and (iii) thermoprotection against heat inactivation of firefly luciferase, and (iv) sequence homology analysis indicated that ICP22 contains an N-terminal J domain and a C-terminal substrate binding domain, similar to type II cellular J proteins. ICP22 may thus be functionally similar to J-protein/Hsp40 cochaperones that function together with their HSP70 partners to prevent aggregation of nonnative proteins. This is not the first example of a virus hijacking a function of a cellular chaperone, since simian immunodeficiency virus T antigen was previously shown to contain a J domain; however, this the first known example of the acquisition of a functional J-like protein by a virus and suggests that HSV has taken advantage of the adaptable nature of J proteins to evolve a multifunctional cochaperone that functions with Hsc70 to promote lytic infection.IMPORTANCE Viruses have evolved a variety of strategies to succeed in a hostile environment. The herpes simplex virus 1 (HSV-1) immediate early protein ICP22 plays several roles in the virus life cycle, including downregulation of cellular gene expression, upregulation of late viral gene expression, inhibition of apoptosis, prevention of aggregation of nonnative proteins, and the recruitment of a cellular heat shock protein, Hsc70, to nuclear domains. We present evidence that ICP22 functionally resembles a cellular J-protein/HSP40 family cochaperone, interacting specifically with Hsc70. We suggest that HSV has taken advantage of the adaptable nature of J proteins to evolve a multifunctional cochaperone that functions with Hsc70 to promote lytic infection.
Collapse
|
42
|
Cdc48/VCP and Endocytosis Regulate TDP-43 and FUS Toxicity and Turnover. Mol Cell Biol 2020; 40:MCB.00256-19. [PMID: 31767634 DOI: 10.1128/mcb.00256-19] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2019] [Accepted: 11/16/2019] [Indexed: 12/14/2022] Open
Abstract
Amyotrophic lateral sclerosis (ALS) is a fatal motor neuron degenerative disease. TDP-43 (TAR DNA-binding protein 43) and FUS (fused in sarcoma) are aggregation-prone RNA-binding proteins that in ALS can mislocalize to the cytoplasm of affected motor neuron cells, often forming cytoplasmic aggregates in the process. Such mislocalization and aggregation are implicated in ALS pathology, though the mechanism(s) of TDP-43 and FUS cytoplasmic toxicity remains unclear. Recently, we determined that the endocytic function aids the turnover (i.e., protein degradation) of TDP-43 and reduces TDP-43 toxicity. Here, we identified that Cdc48 and Ubx3, a Cdc48 cofactor implicated in endocytic function, regulates the turnover and toxicity of TDP-43 and FUS expressed in Saccharomyces cerevisiae Cdc48 physically interacts and colocalizes with TDP-43, as does VCP, in ALS patient tissue. In yeast, FUS toxicity also depends strongly on endocytic function but not on autophagy under normal conditions. FUS expression also impairs endocytic function, as previously observed with TDP-43. Taken together, our data identify a role for Cdc48/VCP and endocytic function in regulating TDP-43 and FUS toxicity and turnover. Furthermore, endocytic dysfunction may be a common defect affecting the cytoplasmic clearance of ALS aggregation-prone proteins and may represent a novel therapeutic target of promise.
Collapse
|
43
|
Jayaraj GG, Hipp MS, Hartl FU. Functional Modules of the Proteostasis Network. Cold Spring Harb Perspect Biol 2020; 12:cshperspect.a033951. [PMID: 30833457 DOI: 10.1101/cshperspect.a033951] [Citation(s) in RCA: 138] [Impact Index Per Article: 27.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cells invest in an extensive network of factors to maintain protein homeostasis (proteostasis) and prevent the accumulation of potentially toxic protein aggregates. This proteostasis network (PN) comprises the machineries for the biogenesis, folding, conformational maintenance, and degradation of proteins with molecular chaperones as central coordinators. Here, we review recent progress in understanding the modular architecture of the PN in mammalian cells and how it is modified during cell differentiation. We discuss the capacity and limitations of the PN in maintaining proteome integrity in the face of proteotoxic stresses, such as aggregate formation in neurodegenerative diseases. Finally, we outline various pharmacological interventions to ameliorate proteostasis imbalance.
Collapse
Affiliation(s)
- Gopal G Jayaraj
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - Mark S Hipp
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| | - F Ulrich Hartl
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Am Klopferspitz 18, 82152 Martinsried, Germany
| |
Collapse
|
44
|
Challenging Proteostasis: Role of the Chaperone Network to Control Aggregation-Prone Proteins in Human Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1243:53-68. [PMID: 32297211 DOI: 10.1007/978-3-030-40204-4_4] [Citation(s) in RCA: 38] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Protein homeostasis (Proteostasis) is essential for correct and efficient protein function within the living cell. Among the critical components of the Proteostasis Network (PN) are molecular chaperones that serve widely in protein biogenesis under physiological conditions, and prevent protein misfolding and aggregation enhanced by conditions of cellular stress. For Alzheimer's, Parkinson's, Huntington's diseases and ALS, multiple classes of molecular chaperones interact with the highly aggregation-prone proteins amyloid-β, tau, α-synuclein, huntingtin and SOD1 to influence the course of proteotoxicity associated with these neurodegenerative diseases. Accordingly, overexpression of molecular chaperones and induction of the heat shock response have been shown to be protective in a wide range of animal models of these diseases. In contrast, for cancer cells the upregulation of chaperones has the undesirable effect of promoting cellular survival and tumor growth by stabilizing mutant oncoproteins. In both situations, physiological levels of molecular chaperones eventually become functionally compromised by the persistence of misfolded substrates, leading to a decline in global protein homeostasis and the dysregulation of diverse cellular pathways. The phenomenon of chaperone competition may underlie the broad pathology observed in aging and neurodegenerative diseases, and restoration of physiological protein homeostasis may be a suitable therapeutic avenue for neurodegeneration as well as for cancer.
Collapse
|
45
|
Ciryam P, Antalek M, Cid F, Tartaglia GG, Dobson CM, Guettsches AK, Eggers B, Vorgerd M, Marcus K, Kley RA, Morimoto RI, Vendruscolo M, Weihl CC. A metastable subproteome underlies inclusion formation in muscle proteinopathies. Acta Neuropathol Commun 2019; 7:197. [PMID: 31796104 PMCID: PMC6891963 DOI: 10.1186/s40478-019-0853-9] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2019] [Accepted: 11/21/2019] [Indexed: 01/20/2023] Open
Abstract
Protein aggregation is a pathological feature of neurodegenerative disorders. We previously demonstrated that protein inclusions in the brain are composed of supersaturated proteins, which are abundant and aggregation-prone, and form a metastable subproteome. It is not yet clear, however, whether this phenomenon is also associated with non-neuronal protein conformational disorders. To respond to this question, we analyzed proteomic datasets from biopsies of patients with genetic and acquired protein aggregate myopathy (PAM) by quantifying the changes in composition, concentration and aggregation propensity of proteins in the fibers containing inclusions and those surrounding them. We found that a metastable subproteome is present in skeletal muscle from healthy patients. The expression of this subproteome escalate as proteomic samples are taken more proximal to the pathologic inclusion, eventually exceeding its solubility limits and aggregating. While most supersaturated proteins decrease or maintain steady abundance across healthy fibers and inclusion-containing fibers, proteins within the metastable subproteome rise in abundance, suggesting that they escape regulation. Taken together, our results show in the context of a human conformational disorder that the supersaturation of a metastable subproteome underlies widespread aggregation and correlates with the histopathological state of the tissue.
Collapse
Affiliation(s)
- Prajwal Ciryam
- Department of Neurology, Vagelos College of Physicians & Surgeons, Columbia University, New York, NY, USA
| | - Matthew Antalek
- Rice Institute for Biomedical Research, Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Fernando Cid
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Gian Gaetano Tartaglia
- Centre for Genomic Regulation, The Barcelona Institute of Science and Technology, Barcelona, Spain
| | - Christopher M Dobson
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Anne-Katrin Guettsches
- Department of Neurology, Heimer Institute of Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Britta Eggers
- Medizinisches Proteom-Center, Ruhr-University Bochum, Bochum, Germany
| | - Matthias Vorgerd
- Department of Neurology, Heimer Institute of Muscle Research, University Hospital Bergmannsheil, Ruhr-University Bochum, Bochum, Germany
| | - Katrin Marcus
- Medizinisches Proteom-Center, Ruhr-University Bochum, Bochum, Germany
| | - Rudolf A Kley
- Department of Neurology, St. Marien Hospital Borken, University of Witten/Herdecke, Borken, Germany
| | - Richard I Morimoto
- Rice Institute for Biomedical Research, Department of Molecular Biosciences, Northwestern University, Evanston, IL, USA
| | - Michele Vendruscolo
- Centre for Misfolding Diseases, Department of Chemistry, University of Cambridge, Cambridge, UK
| | - Conrad C Weihl
- Department of Neurology and Hope Center for Neurological Disease, Washington University School of Medicine, Saint Louis, MO, USA.
| |
Collapse
|
46
|
Abstract
Cells and organisms grow old and die. We develop a biophysical model of the mechanism. Young cells are kept healthy by the positive processes of protein synthesis, degradation, and chaperoning (the activity of keeping proteins properly folded). But, with age, negative processes increase: Oxidative damage accumulates randomly in the cell’s proteins, healthy synthesis and degradation slow down, and—like overfilled garbage cans—chaperone capacity is exceeded. The chaperones are distracted trying to fold irreversibly damaged proteins, leading to accumulating misfolded and aggregated proteins in the cell. The tipping point to death happens when the negative overwhelms the positive. The model makes several quantitative predictions of the life span of the worm Caenorhabditis elegans. What molecular processes drive cell aging and death? Here, we model how proteostasis—i.e., the folding, chaperoning, and maintenance of protein function—collapses with age from slowed translation and cumulative oxidative damage. Irreparably damaged proteins accumulate with age, increasingly distracting the chaperones from folding the healthy proteins the cell needs. The tipping point to death occurs when replenishing good proteins no longer keeps up with depletion from misfolding, aggregation, and damage. The model agrees with experiments in the worm Caenorhabditis elegans that show the following: Life span shortens nonlinearly with increased temperature or added oxidant concentration, and life span increases in mutants having more chaperones or proteasomes. It predicts observed increases in cellular oxidative damage with age and provides a mechanism for the Gompertz-like rise in mortality observed in humans and other organisms. Overall, the model shows how the instability of proteins sets the rate at which damage accumulates with age and upends a cell’s normal proteostasis balance.
Collapse
|
47
|
Da Silva JD, Teixeira-Castro A, Maciel P. From Pathogenesis to Novel Therapeutics for Spinocerebellar Ataxia Type 3: Evading Potholes on the Way to Translation. Neurotherapeutics 2019; 16:1009-1031. [PMID: 31691128 PMCID: PMC6985322 DOI: 10.1007/s13311-019-00798-1] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022] Open
Abstract
Spinocerebellar ataxia type 3 (SCA3), also known as Machado-Joseph disease (MJD), is a neurodegenerative disorder caused by a polyglutamine expansion in the ATXN3 gene. In spite of the identification of a clear monogenic cause 25 years ago, the pathological process still puzzles researchers, impairing prospects for an effective therapy. Here, we propose the disruption of protein homeostasis as the hub of SCA3 pathogenesis, being the molecular mechanisms and cellular pathways that are deregulated in SCA3 downstream consequences of the misfolding and aggregation of ATXN3. Moreover, we attempt to provide a realistic perspective on how the translational/clinical research in SCA3 should evolve. This was based on molecular findings, clinical and epidemiological characteristics, studies of proposed treatments in other conditions, and how that information is essential for their (re-)application in SCA3. This review thus aims i) to critically evaluate the current state of research on SCA3, from fundamental to translational and clinical perspectives; ii) to bring up the current key questions that remain unanswered in this disorder; and iii) to provide a frame on how those answers should be pursued.
Collapse
Affiliation(s)
- Jorge Diogo Da Silva
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Andreia Teixeira-Castro
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal
| | - Patrícia Maciel
- Life and Health Sciences Research Institute (ICVS), School of Medicine, University of Minho, Braga, Portugal.
- ICVS/3B's - PT Government Associate Laboratory, Braga/Guimarães, Portugal.
| |
Collapse
|
48
|
Sitaraman S, Na CL, Yang L, Filuta A, Bridges JP, Weaver TE. Proteasome dysfunction in alveolar type 2 epithelial cells is associated with acute respiratory distress syndrome. Sci Rep 2019; 9:12509. [PMID: 31467330 PMCID: PMC6715642 DOI: 10.1038/s41598-019-49020-4] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2019] [Accepted: 08/19/2019] [Indexed: 01/06/2023] Open
Abstract
Proteasomes are a critical component of quality control that regulate turnover of short-lived, unfolded, and misfolded proteins. Proteasome activity has been therapeutically targeted and considered as a treatment option for several chronic lung disorders including pulmonary fibrosis. Although pharmacologic inhibition of proteasome activity effectively prevents the transformation of fibroblasts to myofibroblasts, the effect on alveolar type 2 (AT2) epithelial cells is not clear. To address this knowledge gap, we generated a genetic model in which a proteasome subunit, RPT3, which promotes assembly of active 26S proteasome, was conditionally deleted in AT2 cells of mice. Partial deletion of RPT3 resulted in 26S proteasome dysfunction, leading to augmented cell stress and cell death. Acute loss of AT2 cells resulted in depletion of alveolar surfactant, disruption of the alveolar epithelial barrier and, ultimately, lethal acute respiratory distress syndrome (ARDS). This study underscores importance of proteasome function in maintenance of AT2 cell homeostasis and supports the need to further investigate the role of proteasome dysfunction in ARDS pathogenesis.
Collapse
Affiliation(s)
- Sneha Sitaraman
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Cheng-Lun Na
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Li Yang
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - Alyssa Filuta
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA
| | - James P Bridges
- Division of Pulmonary, Critical Care and Sleep Medicine, National Jewish Health, Denver, Colorado, 80206, USA
| | - Timothy E Weaver
- Division of Pulmonary Biology, Cincinnati Children's Hospital Medical Center, Cincinnati, Ohio, 45229, USA.
| |
Collapse
|
49
|
Rai SN, Singh BK, Rathore AS, Zahra W, Keswani C, Birla H, Singh SS, Dilnashin H, Singh SP. Quality Control in Huntington's Disease: a Therapeutic Target. Neurotox Res 2019; 36:612-626. [PMID: 31297710 DOI: 10.1007/s12640-019-00087-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2019] [Revised: 07/02/2019] [Accepted: 07/04/2019] [Indexed: 12/19/2022]
Abstract
Huntington's disease (HD) is a fatal autosomal dominantly inherited brain disease caused by excessively expanded CAG repeats in gene which encodes huntingtin protein. These abnormally encoded huntingtin proteins and their truncated fragments result in disruption of cellular quality mechanism ultimately triggering neuronal death. Despite great efforts, a potential causative agent leading to genetic mutation in HTT, manifesting the neurons more prone to oxidative stress, cellular inflammation, energy depletion and apoptotic death, has not been established yet. Current scenario concentrates on symptomatic pathologies to improvise the disease progression and to better the survival. Most of the therapeutic developments have been converged to rescue the protein homeostasis. In HD, abnormal expansion of glutamine repeats in the protein huntingtin leads to toxic aggregation of huntingtin which in turn impairs the quality control mechanism of cells through damaging the machineries involved in removal of aggregated abnormal protein. Therapeutic approaches to improve the efficiency of aggregate clearance through quality control mechanisms involve protein folding machineries such as chaperones and protein degradation machineries such as proteasome and autophagy. Also, to reduce protein aggregation by enhancing proper folding, to degrade and eliminate the aggregates are suggested to negatively regulate the HD progression associated with the disruption of protein homeostasis. This review focuses on the collection of therapeutic strategies targeting enhancement of protein quality control activity to delay the HD pathogenesis.
Collapse
Affiliation(s)
- Sachchida Nand Rai
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Brijesh Kumar Singh
- Department of Pathology and Cell Biology, Columbia University Medical Centre, Columbia University, New York, NY, 10032, USA
| | - Aaina Singh Rathore
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Walia Zahra
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Chetan Keswani
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Hareram Birla
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Saumitra Sen Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Hagera Dilnashin
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India
| | - Surya Pratap Singh
- Department of Biochemistry, Institute of Science, Banaras Hindu University, Varanasi, 221005, India.
| |
Collapse
|
50
|
Wentink A, Nussbaum-Krammer C, Bukau B. Modulation of Amyloid States by Molecular Chaperones. Cold Spring Harb Perspect Biol 2019; 11:a033969. [PMID: 30755450 PMCID: PMC6601462 DOI: 10.1101/cshperspect.a033969] [Citation(s) in RCA: 58] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Aberrant protein aggregation is a defining feature of most neurodegenerative diseases. During pathological aggregation, key proteins transition from their native state to alternative conformations, which are prone to oligomerize into highly ordered fibrillar states. As part of the cellular quality control machinery, molecular chaperones can intervene at many stages of the aggregation process to inhibit or reverse aberrant protein aggregation or counteract the toxicity associated with amyloid species. Although the action of chaperones is considered cytoprotective, essential housekeeping functions can be hijacked for the propagation and spreading of protein aggregates, suggesting the cellular protein quality control system constitutes a double-edged sword in neurodegeneration. Here, we discuss the various mechanisms used by chaperones to influence protein aggregation into amyloid fibrils to understand how the interplay of these activities produces specific cellular outcomes and to define mechanisms that may be targeted by pharmacological agents for the treatment of neurodegenerative conditions.
Collapse
Affiliation(s)
- Anne Wentink
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | - Carmen Nussbaum-Krammer
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| | - Bernd Bukau
- Center for Molecular Biology of Heidelberg University (ZMBH) and German Cancer Research Center (DKFZ), DKFZ-ZMBH Alliance, D-69120 Heidelberg, Germany
| |
Collapse
|