1
|
Stavrides P, Goulbourne CN, Peddy J, Huo C, Rao M, Khetarpal V, Marchionini DM, Nixon RA, Yang DS. mTOR inhibition in Q175 Huntington's disease model mice facilitates neuronal autophagy and mutant huntingtin clearance. eLife 2025; 14:RP104979. [PMID: 40392702 PMCID: PMC12092004 DOI: 10.7554/elife.104979] [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] [Indexed: 05/22/2025] Open
Abstract
Huntington's disease (HD) is caused by the expansion of the polyglutamine stretch in huntingtin protein (HTT) resulting in hallmark aggresomes/inclusion bodies (IBs) composed of mutant huntingtin protein (mHTT) and its fragments. Stimulating autophagy to enhance mHTT clearance is considered a potential therapeutic strategy for HD. Our recent evaluation of the autophagic-lysosomal pathway (ALP) in human HD brain reveals upregulated lysosomal biogenesis and relatively normal autophagy flux in early Vonsattel grade brains, but impaired autolysosome clearance in late grade brains, suggesting that autophagy stimulation could have therapeutic benefits as an early clinical intervention. Here, we tested this hypothesis by crossing the Q175 HD knock-in model with our autophagy reporter mouse TRGL (Thy-1-RFP-GFP-LC3) to investigate in vivo neuronal ALP dynamics. In the Q175 and/or TRGL/Q175 mice, mHTT was detected in autophagic vacuoles and also exhibited a high level of colocalization with autophagy receptors p62/SQSTM1 and ubiquitin in the IBs. Compared to the robust lysosomal pathology in late-stage human HD striatum, ALP alterations in Q175 models are also late-onset but milder, that included a lowered phospho-p70S6K level, lysosome depletion, and autolysosome elevation including more poorly acidified autolysosomes and larger-sized lipofuscin granules, reflecting impaired autophagic flux. Administration of a mTOR inhibitor to 6-mo-old TRGL/Q175 normalized lysosome number, ameliorated aggresome pathology while reducing mHTT-, p62-, and ubiquitin-immunoreactivities, suggesting the beneficial potential of autophagy modulation at early stages of disease progression.
Collapse
Affiliation(s)
- Philip Stavrides
- Center for Dementia Research, Nathan S. Kline InstituteOrangeburgUnited States
| | - Chris N Goulbourne
- Center for Dementia Research, Nathan S. Kline InstituteOrangeburgUnited States
| | - James Peddy
- Center for Dementia Research, Nathan S. Kline InstituteOrangeburgUnited States
| | - Chunfeng Huo
- Center for Dementia Research, Nathan S. Kline InstituteOrangeburgUnited States
| | - Mala Rao
- Center for Dementia Research, Nathan S. Kline InstituteOrangeburgUnited States
- Department of Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
| | | | | | - Ralph A Nixon
- Center for Dementia Research, Nathan S. Kline InstituteOrangeburgUnited States
- Department of Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
- Neuroscience Institute, New York University Grossman School of MedicineNew YorkUnited States
- Department of Cell Biology, New York University Grossman School of MedicineNew YorkUnited States
| | - Dun-Sheng Yang
- Center for Dementia Research, Nathan S. Kline InstituteOrangeburgUnited States
- Department of Psychiatry, New York University Grossman School of MedicineNew YorkUnited States
| |
Collapse
|
2
|
Stavrides P, Goulbourne CN, Peddy J, Huo C, Rao M, Khetarpal V, Marchionini DM, Nixon RA, Yang DS. mTOR inhibition in Q175 Huntington's disease model mice facilitates neuronal autophagy and mutant huntingtin clearance. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.05.29.596471. [PMID: 38854023 PMCID: PMC11160779 DOI: 10.1101/2024.05.29.596471] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Huntington's disease (HD) is caused by expansion of the polyglutamine stretch in huntingtin protein (HTT) resulting in hallmark aggresomes/inclusion bodies (IBs) composed of mutant huntingtin protein (mHTT) and its fragments. Stimulating autophagy to enhance mHTT clearance is considered a potential therapeutic strategy for HD. Our recent evaluation of the autophagic-lysosomal pathway (ALP) in human HD brain reveals upregulated lysosomal biogenesis and relatively normal autophagy flux in early Vonsattel grade brains, but impaired autolysosome clearance in late grade brains, suggesting that autophagy stimulation could have therapeutic benefits as an earlier clinical intervention. Here, we tested this hypothesis by crossing the Q175 HD knock-in model with our autophagy reporter mouse TRGL ( T hy-1- R FP- G FP- L C3) to investigate in vivo neuronal ALP dynamics. In the Q175 and/or TRGL/Q175 mice, mHTT was detected in autophagic vacuoles and also exhibited a high level of colocalization with autophagy receptors p62/SQSTM1 and ubiquitin in the IBs. Compared to the robust lysosomal pathology in late-stage human HD striatum, ALP alterations in Q175 models are also late-onset but milder that included a lowered phospho-p70S6K level, lysosome depletion and autolysosome elevation including more poorly acidified autolysosomes and larger-sized lipofuscin granules, reflecting impaired autophagic flux. Administration of a mTOR inhibitor to 6-mo-old TRGL/Q175 normalized lysosome number, ameliorated aggresome pathology while reducing mHTT-, p62- and ubiquitin-immunoreactivities, suggesting the beneficial potential of autophagy modulation at early stages of disease progression.
Collapse
|
3
|
Cho H. The N17 domain of huntingtin as a multifaceted player in Huntington's disease. Front Mol Biosci 2025; 11:1527313. [PMID: 39845903 PMCID: PMC11753208 DOI: 10.3389/fmolb.2024.1527313] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2024] [Accepted: 12/19/2024] [Indexed: 01/24/2025] Open
Abstract
Huntington's disease (HD) is primarily caused by the aberrant aggregation of the N-terminal exon 1 fragment of mutant huntingtin protein (mHttex1) with expanded polyglutamine (polyQ) repeats in neurons. The first 17 amino acids of the N-terminus of Httex1 (N17 domain) immediately preceding the polyQ repeat domain are evolutionarily conserved across vertebrates and play multifaceted roles in the pathogenesis of HD. Due to its amphipathic helical properties, the N17 domain, both alone and when membrane-associated, promotes mHttEx1 aggregation. Diverse post-translational modifications (PTMs) in the N17 domain alter the aggregation state, thus modulating the cellular toxicity of mHttex1. Furthermore, the N17 domain serves as a nuclear export signal (NES) and mediates the cytoplasmic localization of mHttex1. This review summarizes the four main roles of the N17 domain in regulating HD pathology and discusses potential therapeutic approaches targeting this N17 domain to mitigate HD progression.
Collapse
Affiliation(s)
- Hyunju Cho
- Center for Biomolecular and Cellular Structure, Institute for Basic Science, Daejeon, Republic of Korea
| |
Collapse
|
4
|
Hana TA, Mousa VG, Lin A, Haj-Hussein RN, Michael AH, Aziz MN, Kamaridinova SU, Basnet S, Ormerod KG. Developmental and physiological impacts of pathogenic human huntingtin protein in the nervous system. Neurobiol Dis 2024; 203:106732. [PMID: 39542221 PMCID: PMC12067449 DOI: 10.1016/j.nbd.2024.106732] [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: 09/16/2024] [Revised: 10/29/2024] [Accepted: 11/07/2024] [Indexed: 11/17/2024] Open
Abstract
Huntington's Disease (HD) is a neurodegenerative disorder, part of the nine identified inherited polyglutamine (polyQ) diseases. Most commonly, HD pathophysiology manifests in middle-aged adults with symptoms including progressive loss of motor control, cognitive decline, and psychiatric disturbances. Associated with the pathophysiology of HD is the formation of insoluble fragments of the huntingtin protein (htt) that tend to aggregate in the nucleus and cytoplasm of neurons. To track both the intracellular progression of the aggregation phenotype as well as the physiological deficits associated with mutant htt, two constructs of human HTT were expressed in the Drosophila melanogaster nervous system with varying polyQ lengths, non-pathogenic-htt (NP-htt) and pathogenic-htt (P-htt), with an N-terminal RFP tag for in vivo visualization. P-htt aggregates accumulate in the ventral nerve cord cell bodies as early as 24 h post hatching and significant aggregates form in the segmental nerve branches at 48 h post hatching. Organelle trafficking up- and downstream of aggregates formed in motor neurons showed severe deficits in trafficking dynamics. To explore putative downstream deficits of htt aggregation, ultrastructural changes of presynaptic motor neurons and muscles were assessed, but no significant effects were observed. However, the force and kinetics of muscle contractions were severely affected in P-htt animals, reminiscent of human chorea. Reduced muscle force production translated to altered locomotory behavior. A novel HD aggregation model was established to track htt aggregation throughout adulthood in the wing, showing similar aggregation patterns with larvae. Expressing P-htt in the adult nervous system resulted in significantly reduced lifespan, which could be partially rescued by feeding flies the mTOR inhibitor rapamycin. These findings advance our understanding of htt aggregate progression as well the downstream physiological impacts on the nervous system and peripheral tissues.
Collapse
Affiliation(s)
- Tadros A Hana
- Middle Tennessee State University, Biology Department, Murfreesboro, TN 37132, United States of America
| | - Veronika G Mousa
- Middle Tennessee State University, Biology Department, Murfreesboro, TN 37132, United States of America
| | - Alice Lin
- Brown University, Neuroscience Graduate Program, Warren Alpert Medical School, Providence, RI 02906, United States of America
| | - Rawan N Haj-Hussein
- Middle Tennessee State University, Biology Department, Murfreesboro, TN 37132, United States of America
| | - Andrew H Michael
- Middle Tennessee State University, Biology Department, Murfreesboro, TN 37132, United States of America
| | - Madona N Aziz
- Middle Tennessee State University, Biology Department, Murfreesboro, TN 37132, United States of America
| | - Sevinch U Kamaridinova
- Middle Tennessee State University, Biology Department, Murfreesboro, TN 37132, United States of America
| | - Sabita Basnet
- Middle Tennessee State University, Biology Department, Murfreesboro, TN 37132, United States of America
| | - Kiel G Ormerod
- Middle Tennessee State University, Biology Department, Murfreesboro, TN 37132, United States of America.
| |
Collapse
|
5
|
Hana TA, Mousa VG, Lin A, Haj-Hussein RN, Michael AH, Aziz MN, Kamaridinova SU, Basnet S, Ormerod KG. Developmental and physiological impacts of pathogenic human huntingtin protein in the nervous system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.08.30.610525. [PMID: 39257834 PMCID: PMC11383668 DOI: 10.1101/2024.08.30.610525] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 09/12/2024]
Abstract
Huntington's Disease (HD) is a neurodegenerative disorder, part of the nine identified inherited polyglutamine (polyQ) diseases. Most commonly, HD pathophysiology manifests in middle-aged adults with symptoms including progressive loss of motor control, cognitive decline, and psychiatric disturbances. Associated with the pathophysiology of HD is the formation of insoluble fragments of the huntingtin protein (htt) that tend to aggregate in the nucleus and cytoplasm of neurons. To track both the intracellular progression of the aggregation phenotype as well as the physiological deficits associated with mutant htt, two constructs of human HTT were expressed with varying polyQ lengths, non-pathogenic-htt (Q15, NP-htt) and pathogenic-htt (Q138, P-htt), with an N-terminal RFP tag for in vivo visualization. P-htt aggregates accumulate in the ventral nerve cord cell bodies as early as 24 hours post hatching and significant aggregates form in the segmental nerve branches at 48 hours post hatching. Organelle trafficking up-and downstream of aggregates formed in motor neurons showed severe deficits in trafficking dynamics. To explore putative downstream deficits of htt aggregation, ultrastructural changes of presynaptic motor neurons and muscles were assessed, but no significant effects were observed. However, the force and kinetics of muscle contractions were severely affected in P-htt animals, reminiscent of human chorea. Reduced muscle force production translated to altered locomotory behavior. A novel HD aggregation model was established to track htt aggregation throughout adulthood in the wing, showing similar aggregation patterns with larvae. Expressing P-htt in the adult nervous system resulted in significantly reduced lifespan, which could be partially rescued by feeding flies the mTOR inhibitor rapamycin. These findings advance our understanding of htt aggregate progression as well the downstream physiological impacts on the nervous system and peripheral tissues.
Collapse
|
6
|
Sujkowski A, Ranxhi B, Bangash ZR, Chbihi ZM, Prifti MV, Qadri Z, Alam N, Todi SV, Tsou WL. Progressive degeneration in a new Drosophila model of spinocerebellar ataxia type 7. Sci Rep 2024; 14:14332. [PMID: 38906973 PMCID: PMC11192756 DOI: 10.1038/s41598-024-65172-4] [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: 11/10/2023] [Accepted: 06/18/2024] [Indexed: 06/23/2024] Open
Abstract
Spinocerebellar ataxia type 7 (SCA7) is a progressive neurodegenerative disorder resulting from abnormal expansion of an uninterrupted polyglutamine (polyQ) repeat in its disease protein, ataxin-7 (ATXN7). ATXN7 is part of Spt-Ada-Gcn5 acetyltransferase (SAGA), an evolutionarily conserved transcriptional coactivation complex with critical roles in chromatin remodeling, cell signaling, neurodifferentiation, mitochondrial health and autophagy. SCA7 is dominantly inherited and characterized by genetic anticipation and high repeat-length instability. Patients with SCA7 experience progressive ataxia, atrophy, spasticity, and blindness. There is currently no cure for SCA7, and therapies are aimed at alleviating symptoms to increase quality of life. Here, we report novel Drosophila lines of SCA7 with polyQ repeats in wild-type and human disease patient range. We find that ATXN7 expression has age- and polyQ repeat length-dependent reduction in fruit fly survival and retinal instability, concomitant with increased ATXN7 protein aggregation. These new lines will provide important insight on disease progression that can be used in the future to identify therapeutic targets for SCA7 patients.
Collapse
Affiliation(s)
- Alyson Sujkowski
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA
| | - Bedri Ranxhi
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA
| | - Zoya R Bangash
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA
| | - Zachary M Chbihi
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA
| | - Matthew V Prifti
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA
| | - Zaina Qadri
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA
| | - Nadir Alam
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA
| | - Sokol V Todi
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA
- Department of Neurology, Wayne State University School of Medicine, Detroit, MI, USA
| | - Wei-Ling Tsou
- Department of Pharmacology, Wayne State University School of Medicine, 540 E Canfield, Scott Hall Rm 3108, Detroit, MI, 48201, USA.
| |
Collapse
|
7
|
Shafie A, Ashour AA, Anwar S, Anjum F, Hassan MI. Exploring molecular mechanisms, therapeutic strategies, and clinical manifestations of Huntington's disease. Arch Pharm Res 2024; 47:571-595. [PMID: 38764004 DOI: 10.1007/s12272-024-01499-w] [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: 12/18/2023] [Accepted: 05/02/2024] [Indexed: 05/21/2024]
Abstract
Huntington's disease (HD) is a paradigm of a genetic neurodegenerative disorder characterized by the expansion of CAG repeats in the HTT gene. This extensive review investigates the molecular complexities of HD by highlighting the pathogenic mechanisms initiated by the mutant huntingtin protein. Adverse outcomes of HD include mitochondrial dysfunction, compromised protein clearance, and disruption of intracellular signaling, consequently contributing to the gradual deterioration of neurons. Numerous therapeutic strategies, particularly precision medicine, are currently used for HD management. Antisense oligonucleotides, such as Tominersen, play a leading role in targeting and modulating the expression of mutant huntingtin. Despite the promise of these therapies, challenges persist, particularly in improving delivery systems and the necessity for long-term safety assessments. Considering the future landscape, the review delineates promising directions for HD research and treatment. Innovations such as Clustered regularly interspaced short palindromic repeats associated system therapies (CRISPR)-based genome editing and emerging neuroprotective approaches present unprecedented opportunities for intervention. Collaborative interdisciplinary endeavors and a more insightful understanding of HD pathogenesis are on the verge of reshaping the therapeutic landscape. As we navigate the intricate landscape of HD, this review serves as a guide for unraveling the intricacies of this disease and progressing toward transformative treatments.
Collapse
Affiliation(s)
- Alaa Shafie
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, PO Box 11099, 21944, Taif, Saudi Arabia
| | - Amal Adnan Ashour
- Department of Oral and Maxillofacial Surgery and Diagnostic Sciences, Faculty of Dentistry, Taif University, PO Box 11099, 21944, Taif, Saudi Arabia
| | - Saleha Anwar
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India
| | - Farah Anjum
- Department of Clinical Laboratory Sciences, College of Applied Medical Sciences, Taif University, PO Box 11099, 21944, Taif, Saudi Arabia
| | - Md Imtaiyaz Hassan
- Centre for Interdisciplinary Research in Basic Sciences, Jamia Millia Islamia, Jamia Nagar, New Delhi, 110025, India.
| |
Collapse
|
8
|
Berg MJ, Veeranna, Rosa CM, Kumar A, Mohan PS, Stavrides P, Marchionini DM, Yang DS, Nixon RA. Pathobiology of the autophagy-lysosomal pathway in the Huntington's disease brain. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.05.29.596470. [PMID: 38854113 PMCID: PMC11160756 DOI: 10.1101/2024.05.29.596470] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2024]
Abstract
Accumulated levels of mutant huntingtin protein (mHTT) and its fragments are considered contributors to the pathogenesis of Huntington's disease (HD). Although lowering mHTT by stimulating autophagy has been considered a possible therapeutic strategy, the role and competence of autophagy-lysosomal pathway (ALP) during HD progression in the human disease remains largely unknown. Here, we used multiplex confocal and ultrastructural immunocytochemical analyses of ALP functional markers in relation to mHTT aggresome pathology in striatum and the less affected cortex of HD brains staged from HD2 to HD4 by Vonsattel neuropathological criteria compared to controls. Immunolabeling revealed the localization of HTT/mHTT in ALP vesicular compartments labeled by autophagy-related adaptor proteins p62/SQSTM1 and ubiquitin, and cathepsin D (CTSD) as well as HTT-positive inclusions. Although comparatively normal at HD2, neurons at later HD stages exhibited progressive enlargement and clustering of CTSD-immunoreactive autolysosomes/lysosomes and, ultrastructurally, autophagic vacuole/lipofuscin granules accumulated progressively, more prominently in striatum than cortex. These changes were accompanied by rises in levels of HTT/mHTT and p62/SQSTM1, particularly their fragments, in striatum but not in the cortex, and by increases of LAMP1 and LAMP2 RNA and LAMP1 protein. Importantly, no blockage in autophagosome formation and autophagosome-lysosome fusion was detected, thus pinpointing autophagy substrate clearance deficits as a basis for autophagic flux declines. The findings collectively suggest that upregulated lysosomal biogenesis and preserved proteolysis maintain autophagic clearance in early-stage HD, but failure at advanced stages contributes to progressive HTT build-up and potential neurotoxicity. These findings support the prospect that ALP stimulation applied at early disease stages, when clearance machinery is fully competent, may have therapeutic benefits in HD patients.
Collapse
|
9
|
Tong H, Yang T, Xu S, Li X, Liu L, Zhou G, Yang S, Yin S, Li XJ, Li S. Huntington's Disease: Complex Pathogenesis and Therapeutic Strategies. Int J Mol Sci 2024; 25:3845. [PMID: 38612657 PMCID: PMC11011923 DOI: 10.3390/ijms25073845] [Citation(s) in RCA: 18] [Impact Index Per Article: 18.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2024] [Revised: 03/21/2024] [Accepted: 03/26/2024] [Indexed: 04/14/2024] Open
Abstract
Huntington's disease (HD) arises from the abnormal expansion of CAG repeats in the huntingtin gene (HTT), resulting in the production of the mutant huntingtin protein (mHTT) with a polyglutamine stretch in its N-terminus. The pathogenic mechanisms underlying HD are complex and not yet fully elucidated. However, mHTT forms aggregates and accumulates abnormally in neuronal nuclei and processes, leading to disruptions in multiple cellular functions. Although there is currently no effective curative treatment for HD, significant progress has been made in developing various therapeutic strategies to treat HD. In addition to drugs targeting the neuronal toxicity of mHTT, gene therapy approaches that aim to reduce the expression of the mutant HTT gene hold great promise for effective HD therapy. This review provides an overview of current HD treatments, discusses different therapeutic strategies, and aims to facilitate future therapeutic advancements in the field.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Xiao-Jiang Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (H.T.); (T.Y.); (S.X.); (X.L.); (L.L.); (G.Z.); (S.Y.); (S.Y.)
| | - Shihua Li
- Guangdong Key Laboratory of Non-Human Primate Research, Key Laboratory of CNS Regeneration (Ministry of Education), Guangdong-Hongkong-Macau Institute of CNS Regeneration, Jinan University, Guangzhou 510632, China; (H.T.); (T.Y.); (S.X.); (X.L.); (L.L.); (G.Z.); (S.Y.); (S.Y.)
| |
Collapse
|
10
|
Geijtenbeek KW, Aranda AS, Sanz AS, Janzen J, Bury AE, Kors S, Al Amery N, Schmitz NCM, Reits EAJ, Schipper-Krom S. Insulin-Degrading Enzyme Efficiently Degrades polyQ Peptides but not Expanded polyQ Huntingtin Fragments. J Huntingtons Dis 2024; 13:201-214. [PMID: 38640164 DOI: 10.3233/jhd-230583] [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] [Indexed: 04/21/2024]
Abstract
Background Huntington's disease is an inheritable autosomal dominant disorder caused by an expanded CAG trinucleotide repeat within the Huntingtin gene, leading to a polyglutamine (polyQ) expansion in the mutant protein. Objective A potential therapeutic approach for delaying or preventing the onset of the disease involves enhancing the degradation of the aggregation-prone polyQ-expanded N-terminal mutant huntingtin (mHTT) exon1 fragment. A few proteases and peptidases have been identified that are able to cleave polyQ fragments with low efficiency. This study aims to identify a potent polyQ-degrading endopeptidase. Methods Here we used quenched polyQ peptides to identify a polyQ-degrading endopeptidase. Next we investigated its role on HTT turnover, using purified polyQ-expanded HTT fragments and striatal cells expressing mHTT exon1 peptides. Results We identified insulin-degrading enzyme (IDE) as a novel endopeptidase for degrading polyQ peptides. IDE was, however, ineffective in reducing purified polyQ-expanded HTT fragments. Similarly, in striatal cells expressing mHTT exon1 peptides, IDE did not enhance mHTT turnover. Conclusions This study shows that despite IDE's efficiency in degrading polyQ peptides, it does not contribute to the direct degradation of polyQ-expanded mHTT fragments.
Collapse
Affiliation(s)
- Karlijne W Geijtenbeek
- Amsterdam UMC, University of Amsterdam, Medical Biology, Meibergdreef, Amsterdam, Netherlands
| | - Angela Santiago Aranda
- Amsterdam UMC, University of Amsterdam, Medical Biology, Meibergdreef, Amsterdam, Netherlands
| | - Alicia Sanz Sanz
- Amsterdam UMC, University of Amsterdam, Medical Biology, Meibergdreef, Amsterdam, Netherlands
| | - Jolien Janzen
- Amsterdam UMC, University of Amsterdam, Medical Biology, Meibergdreef, Amsterdam, Netherlands
| | - Aleksandra E Bury
- Amsterdam UMC, University of Amsterdam, Medical Biology, Meibergdreef, Amsterdam, Netherlands
| | - Suzan Kors
- Amsterdam UMC, University of Amsterdam, Medical Biology, Meibergdreef, Amsterdam, Netherlands
| | | | - Nina C M Schmitz
- Amsterdam UMC, University of Amsterdam, Medical Biology, Meibergdreef, Amsterdam, Netherlands
| | - Eric A J Reits
- Amsterdam UMC, University of Amsterdam, Medical Biology, Meibergdreef, Amsterdam, Netherlands
| | - Sabine Schipper-Krom
- Amsterdam UMC, University of Amsterdam, Medical Biology, Meibergdreef, Amsterdam, Netherlands
| |
Collapse
|
11
|
Zhang H, Wu S, Itzhaki LS, Perrett S. Interaction between huntingtin exon 1 and HEAT repeat structure probed by chimeric model proteins. Protein Sci 2023; 32:e4810. [PMID: 37853955 PMCID: PMC10659953 DOI: 10.1002/pro.4810] [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: 06/29/2023] [Revised: 10/11/2023] [Accepted: 10/15/2023] [Indexed: 10/20/2023]
Abstract
Huntington disease (HD) is associated with aggregation of huntingtin (HTT) protein containing over 35 continuous Q residues within the N-terminal exon 1 encoded region. The C-terminal of the HTT protein consists mainly of HEAT repeat structure which serves as a scaffold for multiple cellular activities. Structural and biochemical analysis of the intact HTT protein has been hampered by its huge size (~300 kDa) and most in vitro studies to date have focused on the properties of the exon 1 region. To explore the interaction between HTT exon 1 and the HEAT repeat structure, we constructed chimeric proteins containing the N-terminal HTT exon 1 region and the HEAT repeat protein PR65/A. The results indicate that HTT exon 1 slightly destabilizes the downstream HEAT repeat structure and endows the HEAT repeat structure with more conformational flexibility. Wild-type and pathological lengths of polyQ did not show differences in the interaction between HTT exon 1 and the HEAT repeats. With the C-terminal fusion of PR65/A, HTT exon 1 containing pathological lengths of polyQ could still form amyloid fibrils, but the higher-order architecture of fibrils and kinetics of fibril formation were affected by the C-terminal fusion of HEAT repeats. This indicates that interaction between HTT exon 1 and HEAT repeat structure is compatible with both normal function of HTT protein and the pathogenesis of HD, and this study provides a potential model for further exploration.
Collapse
Affiliation(s)
- Hong Zhang
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- University of the Chinese Academy of SciencesBeijingChina
- Institute of Basic Medical Sciences, Chinese Academy of Medical SciencesPeking Union Medical CollegeBeijingChina
| | - Si Wu
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- University of the Chinese Academy of SciencesBeijingChina
| | | | - Sarah Perrett
- National Laboratory of Biomacromolecules, CAS Center for Excellence in BiomacromoleculesInstitute of Biophysics, Chinese Academy of SciencesBeijingChina
- University of the Chinese Academy of SciencesBeijingChina
| |
Collapse
|
12
|
Sujkowski AL, Ranxhi B, Prifti MV, Alam N, Todi SV, Tsou WL. Progressive degeneration in a new Drosophila model of Spinocerebellar Ataxia type 7. RESEARCH SQUARE 2023:rs.3.rs-3592641. [PMID: 38045332 PMCID: PMC10690306 DOI: 10.21203/rs.3.rs-3592641/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/05/2023]
Abstract
Spinocerebellar ataxia type 7 (SCA7) is a progressive neurodegenerative disorder resulting from abnormal expansion of polyglutamine (polyQ) in its disease protein, ataxin-7 (ATXN7). ATXN7 is part of Spt-Ada-Gcn5 acetyltransferase (SAGA), an evolutionarily conserved transcriptional coactivation complex with critical roles in chromatin remodeling, cell signaling, neurodifferentiation, mitochondrial health and autophagy. SCA7 is dominantly inherited and characterized by genetic anticipation and high repeat-length instability. Patients with SCA7 experience progressive ataxia, atrophy, spasticity, and blindness. There is currently no cure for SCA7, and therapies are aimed at alleviating symptoms to increase quality of life. Here, we report novel Drosophila lines of SCA7 with polyQ repeats in wild-type and human disease patient range. We find that ATXN7 expression has age- and polyQ repeat length-dependent reduction in survival and retinal instability, concomitant with increased ATXN7 protein aggregation. These new lines will provide important insight on disease progression that can be used in the future to identify therapeutic targets for SCA7 patients.
Collapse
Affiliation(s)
| | - Bedri Ranxhi
- Department of Pharmacology, Wayne State University School of Medicine
| | - Matthew V Prifti
- Department of Pharmacology, Wayne State University School of Medicine
| | - Nadir Alam
- Department of Pharmacology, Wayne State University School of Medicine
| | - Sokol V Todi
- Department of Pharmacology, Wayne State University School of Medicine
- Department of Neurology, Wayne State University School of Medicine
| | - Wei-Ling Tsou
- Department of Pharmacology, Wayne State University School of Medicine
| |
Collapse
|
13
|
Sujkowski AL, Ranxhi B, Prifti MV, Alam N, Todi SV, Tsou WL. Progressive degeneration in a new Drosophila model of Spinocerebellar Ataxia type 7. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.11.07.566106. [PMID: 37986914 PMCID: PMC10659390 DOI: 10.1101/2023.11.07.566106] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2023]
Abstract
Spinocerebellar ataxia type 7 (SCA7) is a progressive neurodegenerative disorder resulting from abnormal expansion of polyglutamine (polyQ) in its disease protein, ataxin-7 (ATXN7). ATXN7 is part of Spt-Ada-Gcn5 acetyltransferase (SAGA), an evolutionarily conserved transcriptional coactivation complex with critical roles in chromatin remodeling, cell signaling, neurodifferentiation, mitochondrial health and autophagy. SCA7 is dominantly inherited and characterized by genetic anticipation and high repeat-length instability. Patients with SCA7 experience progressive ataxia, atrophy, spasticity, and blindness. There is currently no cure for SCA7, and therapies are aimed at alleviating symptoms to increase quality of life. Here, we report novel Drosophila lines of SCA7 with polyQ repeats in wild-type and human disease patient range. We find that ATXN7 expression has age- and polyQ repeat length-dependent reduction in survival and retinal instability, concomitant with increased ATXN7 protein aggregation. These new lines will provide important insight on disease progression that can be used in the future to identify therapeutic targets for SCA7 patients.
Collapse
|
14
|
Reed AL, Mitchell W, Alexandrescu AT, Alder NN. Interactions of amyloidogenic proteins with mitochondrial protein import machinery in aging-related neurodegenerative diseases. Front Physiol 2023; 14:1263420. [PMID: 38028797 PMCID: PMC10652799 DOI: 10.3389/fphys.2023.1263420] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/19/2023] [Accepted: 10/02/2023] [Indexed: 12/01/2023] Open
Abstract
Most mitochondrial proteins are targeted to the organelle by N-terminal mitochondrial targeting sequences (MTSs, or "presequences") that are recognized by the import machinery and subsequently cleaved to yield the mature protein. MTSs do not have conserved amino acid compositions, but share common physicochemical properties, including the ability to form amphipathic α-helical structures enriched with basic and hydrophobic residues on alternating faces. The lack of strict sequence conservation implies that some polypeptides can be mistargeted to mitochondria, especially under cellular stress. The pathogenic accumulation of proteins within mitochondria is implicated in many aging-related neurodegenerative diseases, including Alzheimer's, Parkinson's, and Huntington's diseases. Mechanistically, these diseases may originate in part from mitochondrial interactions with amyloid-β precursor protein (APP) or its cleavage product amyloid-β (Aβ), α-synuclein (α-syn), and mutant forms of huntingtin (mHtt), respectively, that are mediated in part through their associations with the mitochondrial protein import machinery. Emerging evidence suggests that these amyloidogenic proteins may present cryptic targeting signals that act as MTS mimetics and can be recognized by mitochondrial import receptors and transported into different mitochondrial compartments. Accumulation of these mistargeted proteins could overwhelm the import machinery and its associated quality control mechanisms, thereby contributing to neurological disease progression. Alternatively, the uptake of amyloidogenic proteins into mitochondria may be part of a protein quality control mechanism for clearance of cytotoxic proteins. Here we review the pathomechanisms of these diseases as they relate to mitochondrial protein import and effects on mitochondrial function, what features of APP/Aβ, α-syn and mHtt make them suitable substrates for the import machinery, and how this information can be leveraged for the development of therapeutic interventions.
Collapse
Affiliation(s)
- Ashley L. Reed
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, United States
| | - Wayne Mitchell
- Department of Medicine, Brigham and Women’s Hospital, Harvard Medical School, Boston, MA, United States
| | - Andrei T. Alexandrescu
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, United States
| | - Nathan N. Alder
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, CT, United States
| |
Collapse
|
15
|
de Wet S, Theart R, Loos B. Cogs in the autophagic machine-equipped to combat dementia-prone neurodegenerative diseases. Front Mol Neurosci 2023; 16:1225227. [PMID: 37720551 PMCID: PMC10500130 DOI: 10.3389/fnmol.2023.1225227] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2023] [Accepted: 07/31/2023] [Indexed: 09/19/2023] Open
Abstract
Neurodegenerative diseases are often characterized by hydrophobic inclusion bodies, and it may be the case that the aggregate-prone proteins that comprise these inclusion bodies are in fact the cause of neurotoxicity. Indeed, the appearance of protein aggregates leads to a proteostatic imbalance that causes various interruptions in physiological cellular processes, including lysosomal and mitochondrial dysfunction, as well as break down in calcium homeostasis. Oftentimes the approach to counteract proteotoxicity is taken to merely upregulate autophagy, measured by an increase in autophagosomes, without a deeper assessment of contributors toward effective turnover through autophagy. There are various ways in which autophagy is regulated ranging from the mammalian target of rapamycin (mTOR) to acetylation status of proteins. Healthy mitochondria and the intracellular energetic charge they preserve are key for the acidification status of lysosomes and thus ensuring effective clearance of components through the autophagy pathway. Both mitochondria and lysosomes have been shown to bear functional protein complexes that aid in the regulation of autophagy. Indeed, it may be the case that minimizing the proteins associated with the respective neurodegenerative pathology may be of greater importance than addressing molecularly their resulting inclusion bodies. It is in this context that this review will dissect the autophagy signaling pathway, its control and the manner in which it is molecularly and functionally connected with the mitochondrial and lysosomal system, as well as provide a summary of the role of autophagy dysfunction in driving neurodegenerative disease as a means to better position the potential of rapamycin-mediated bioactivities to control autophagy favorably.
Collapse
Affiliation(s)
- Sholto de Wet
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| | - Rensu Theart
- Department of Electric and Electronic Engineering, Stellenbosch University, Stellenbosch, South Africa
| | - Ben Loos
- Department of Physiological Sciences, Stellenbosch University, Stellenbosch, South Africa
| |
Collapse
|
16
|
Nouri Nojadeh J, Bildiren Eryilmaz NS, Ergüder BI. CRISPR/Cas9 genome editing for neurodegenerative diseases. EXCLI JOURNAL 2023; 22:567-582. [PMID: 37636024 PMCID: PMC10450213 DOI: 10.17179/excli2023-6155] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Subscribe] [Scholar Register] [Received: 05/06/2023] [Accepted: 06/29/2023] [Indexed: 08/29/2023]
Abstract
Gene therapy has emerged as a promising therapeutic strategy for various conditions, including blood disorders, ocular disease, cancer, and nervous system disorders. The advent of gene editing techniques has facilitated the ability of researchers to specifically target and modify the eukaryotic cell genome, making it a valuable tool for gene therapy. This can be performed through either in vivo or ex vivo approaches. Gene editing tools, such as zinc finger nucleases, transcription activator-like effector nucleases, and CRISPR-Cas-associated nucleases, can be employed for gene therapy purposes. Among these tools, CRISPR-Cas-based gene editing stands out because of its ability to introduce heritable genome changes by designing short guide RNAs. This review aims to provide an overview of CRISPR-Cas technology and summarizes the latest research on the application of CRISPR/Cas9 genome editing technology for the treatment of the most prevalent neurodegenerative diseases including Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic lateral sclerosis, and Spinocerebellar ataxia.
Collapse
Affiliation(s)
- Jafar Nouri Nojadeh
- Ankara University Faculty of Medicine, Department of Medical Biochemistry, Ankara, Turkey
- The Graduate School of Health Sciences of Ankara University, Ankara, Turkey
| | | | - Berrin Imge Ergüder
- Ankara University Faculty of Medicine, Department of Medical Biochemistry, Ankara, Turkey
- The Graduate School of Health Sciences of Ankara University, Ankara, Turkey
| |
Collapse
|
17
|
Incebacak Eltemur RD, Nguyen HP, Weber JJ. Calpain-mediated proteolysis as driver and modulator of polyglutamine toxicity. Front Mol Neurosci 2022; 15:1020104. [PMID: 36385755 PMCID: PMC9648470 DOI: 10.3389/fnmol.2022.1020104] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2022] [Accepted: 09/26/2022] [Indexed: 09/22/2023] Open
Abstract
Among posttranslational modifications, directed proteolytic processes have the strongest impact on protein integrity. They are executed by a variety of cellular machineries and lead to a wide range of molecular consequences. Compared to other forms of proteolytic enzymes, the class of calcium-activated calpains is considered as modulator proteases due to their limited proteolytic activity, which changes the structure and function of their target substrates. In the context of neurodegeneration and - in particular - polyglutamine disorders, proteolytic events have been linked to modulatory effects on the molecular pathogenesis by generating harmful breakdown products of disease proteins. These findings led to the formulation of the toxic fragment hypothesis, and calpains appeared to be one of the key players and auspicious therapeutic targets in Huntington disease and Machado Joseph disease. This review provides a current survey of the role of calpains in proteolytic processes found in polyglutamine disorders. Together with insights into general concepts behind toxic fragments and findings in polyglutamine disorders, this work aims to inspire researchers to broaden and deepen the knowledge in this field, which will help to evaluate calpain-mediated proteolysis as a unifying and therapeutically targetable posttranslational mechanism in neurodegeneration.
Collapse
Affiliation(s)
- Rana Dilara Incebacak Eltemur
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| | - Huu Phuc Nguyen
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
| | - Jonasz Jeremiasz Weber
- Department of Human Genetics, Ruhr University Bochum, Bochum, Germany
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Tübingen, Germany
| |
Collapse
|
18
|
van der Bent ML, Evers MM, Vallès A. Emerging Therapies for Huntington's Disease - Focus on N-Terminal Huntingtin and Huntingtin Exon 1. Biologics 2022; 16:141-160. [PMID: 36213816 PMCID: PMC9532260 DOI: 10.2147/btt.s270657] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Accepted: 09/14/2022] [Indexed: 11/12/2022]
Abstract
Huntington's disease is a devastating heritable neurodegenerative disorder that is caused by the presence of a trinucleotide CAG repeat expansion in the Huntingtin gene, leading to a polyglutamine tract in the protein. Various mechanisms lead to the production of N-terminal Huntingtin protein fragments, which are reportedly more toxic than the full-length protein. In this review, we summarize the current knowledge on the production and toxicity of N-terminal Huntingtin protein fragments. Further, we expand on various therapeutic strategies targeting N-terminal Huntingtin on the protein, RNA and DNA level. Finally, we compare the therapeutic approaches that are clinically most advanced, including those that do not target N-terminal Huntingtin, discussing differences in mode of action and translational applicability.
Collapse
Affiliation(s)
| | - Melvin M Evers
- uniQure biopharma B.V., Department of Research and Development, Amsterdam, the Netherlands
| | - Astrid Vallès
- uniQure biopharma B.V., Department of Research and Development, Amsterdam, the Netherlands
| |
Collapse
|
19
|
Johnson SL, Tsou WL, Prifti MV, Harris AL, Todi SV. A survey of protein interactions and posttranslational modifications that influence the polyglutamine diseases. Front Mol Neurosci 2022; 15:974167. [PMID: 36187346 PMCID: PMC9515312 DOI: 10.3389/fnmol.2022.974167] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2022] [Accepted: 07/27/2022] [Indexed: 01/20/2023] Open
Abstract
The presence and aggregation of misfolded proteins has deleterious effects in the nervous system. Among the various diseases caused by misfolded proteins is the family of the polyglutamine (polyQ) disorders. This family comprises nine members, all stemming from the same mutation—the abnormal elongation of a polyQ repeat in nine different proteins—which causes protein misfolding and aggregation, cellular dysfunction and disease. While it is the same type of mutation that causes them, each disease is distinct: it is influenced by regions and domains that surround the polyQ repeat; by proteins with which they interact; and by posttranslational modifications they receive. Here, we overview the role of non-polyQ regions that control the pathogenicity of the expanded polyQ repeat. We begin by introducing each polyQ disease, the genes affected, and the symptoms experienced by patients. Subsequently, we provide a survey of protein-protein interactions and posttranslational modifications that regulate polyQ toxicity. We conclude by discussing shared processes and pathways that bring some of the polyQ diseases together and may serve as common therapeutic entry points for this family of incurable disorders.
Collapse
Affiliation(s)
- Sean L. Johnson
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
| | - Wei-Ling Tsou
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
| | - Matthew V. Prifti
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
| | - Autumn L. Harris
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
- Maximizing Access to Research Careers (MARC) Program, Wayne State University, Detroit, MI, United States
| | - Sokol V. Todi
- Department of Pharmacology, Wayne State University, Detroit, MI, United States
- Maximizing Access to Research Careers (MARC) Program, Wayne State University, Detroit, MI, United States
- Department of Neurology, Wayne State University, Detroit, MI, United States
- *Correspondence: Sokol V. Todi,
| |
Collapse
|
20
|
Wu X, Wan T, Gao X, Fu M, Duan Y, Shen X, Guo W. Microglia Pyroptosis: A Candidate Target for Neurological Diseases Treatment. Front Neurosci 2022; 16:922331. [PMID: 35937897 PMCID: PMC9354884 DOI: 10.3389/fnins.2022.922331] [Citation(s) in RCA: 28] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/17/2022] [Accepted: 06/23/2022] [Indexed: 11/13/2022] Open
Abstract
In addition to its profound implications in the fight against cancer, pyroptosis have important role in the regulation of neuronal injury. Microglia are not only central members of the immune regulation of the central nervous system (CNS), but are also involved in the development and homeostatic maintenance of the nervous system. Under various pathological overstimulation, microglia pyroptosis contributes to the massive release of intracellular inflammatory mediators leading to neuroinflammation and ultimately to neuronal damages. In addition, microglia pyroptosis lead to further neurological damage by decreasing the ability to cleanse harmful substances. The pathogenic roles of microglia in a variety of CNS diseases such as neurodegenerative diseases, stroke, multiple sclerosis and depression, and many other neurological disorders have been gradually unveiled. In the context of different neurological disorders, inhibition of microglia pyroptosis by targeting NOD-like receptor family pyrin domain containing (NLRP) 3, caspase-1 and gasdermins (GSDMs) by various chemical agents as well as natural products significantly improve the symptoms or outcome in animal models. This study will provide new ideas for immunomodulatory treatment of CNS diseases.
Collapse
Affiliation(s)
- Xian Wu
- The First Affiliated Hospital of Hunan College of Traditional Chinese Medicine, Hunan Province Directly Affiliated TCM Hospital, Zhuzhou, China
| | - Teng Wan
- Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
| | - Xiaoyu Gao
- Hengyang Medical College, University of South China, Hengyang, China
| | - Mingyuan Fu
- Hengyang Medical College, University of South China, Hengyang, China
| | - Yunfeng Duan
- The First Affiliated Hospital of Hunan College of Traditional Chinese Medicine, Hunan Province Directly Affiliated TCM Hospital, Zhuzhou, China
| | - Xiangru Shen
- Hengyang Medical College, University of South China, Hengyang, China
- *Correspondence: Xiangru Shen
| | - Weiming Guo
- Huazhong University of Science and Technology Union Shenzhen Hospital, The 6th Affiliated Hospital of Shenzhen University Health Science Center, Shenzhen, China
- Weiming Guo
| |
Collapse
|
21
|
Saha S, Mohanta S, Das R, Dalai R, Divyanshi, Tiwari N, Tiwari A, Kumar A, Goswami C. Ratio of Hydrophobic-Hydrophilic and Positive-Negative Residues at Lipid-Water-Interface Influences Surface Expression and Channel Gating of TRPV1. J Membr Biol 2022; 255:319-339. [PMID: 35608627 DOI: 10.1007/s00232-022-00243-z] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2022] [Accepted: 05/03/2022] [Indexed: 12/19/2022]
Abstract
During evolution, TRPV1 has lost, retained or selected certain residues at Lipid-Water-Interface (LWI) and formed specific patterns there. The ratio of "hydrophobic-hydrophilic" and "positive-negative-charged" residues at the inner LWI remains conserved throughout vertebrate evolution and plays important role in regulating TRPV1 trafficking and localization. Arg575 is an important residue as Arg575Asp mutant has reduced surface expression, co-localization with lipid raft markers, cell area and increased cell lethality. This lethality is most likely due to the disruption of the ratio between positive-negative charges caused by the mutation. Such lethality can be rescued by either using TRPV1-specfic inhibitor 5'-IRTX or by restoring the positive-negative charge ratio at that position, i.e. by introducing Asp576Arg mutation in Arg575Asp backbone. We propose that Arg575Asp mutation confers TRPV1 in a "constitutive-open-like" condition. These findings have broader implication in understanding the molecular evolution of thermo-sensitive ion channels and the micro-environments involved in processes that goes erratic in different diseases. The segment of TRPV1 that is present at the inner lipid-water-interface (LWI) has a specific pattern of amino acid combinations. The overall ratio of +ve charge /-ve charge and the ratio of hydrophobicity/hydrophilicity remain constant throughout the vertebrate evolution (ca 450 million years). This specific pattern is not observed in the outer LWI region of TRPV1.
Collapse
Affiliation(s)
- Somdatta Saha
- School of Biological Sciences, National Institute of Science Education and Research, Jatni Campus, Bhubaneswar, Orissa, 752050, India.,Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400094, India
| | - Sushama Mohanta
- School of Biological Sciences, National Institute of Science Education and Research, Jatni Campus, Bhubaneswar, Orissa, 752050, India.,Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400094, India
| | - Rashmita Das
- School of Biological Sciences, National Institute of Science Education and Research, Jatni Campus, Bhubaneswar, Orissa, 752050, India.,Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400094, India
| | - Ritesh Dalai
- School of Biological Sciences, National Institute of Science Education and Research, Jatni Campus, Bhubaneswar, Orissa, 752050, India.,Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400094, India
| | - Divyanshi
- School of Biological Sciences, National Institute of Science Education and Research, Jatni Campus, Bhubaneswar, Orissa, 752050, India.,Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400094, India
| | - Nikhil Tiwari
- School of Biological Sciences, National Institute of Science Education and Research, Jatni Campus, Bhubaneswar, Orissa, 752050, India.,Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400094, India
| | - Ankit Tiwari
- School of Biological Sciences, National Institute of Science Education and Research, Jatni Campus, Bhubaneswar, Orissa, 752050, India
| | - Abhishek Kumar
- Institute of Bioinformatics, International Technology Park, Bangalore, 560066, India.,Manipal Academy of Higher Education (MAHE), Manipal, Karnataka, 576104, India
| | - Chandan Goswami
- School of Biological Sciences, National Institute of Science Education and Research, Jatni Campus, Bhubaneswar, Orissa, 752050, India. .,Homi Bhabha National Institute, Training School Complex, Anushakti Nagar, Mumbai, 400094, India.
| |
Collapse
|
22
|
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: 51] [Impact Index Per Article: 17.0] [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
|
23
|
Moretto E, Stuart S, Surana S, Vargas JNS, Schiavo G. The Role of Extracellular Matrix Components in the Spreading of Pathological Protein Aggregates. Front Cell Neurosci 2022; 16:844211. [PMID: 35573838 PMCID: PMC9100790 DOI: 10.3389/fncel.2022.844211] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2021] [Accepted: 03/08/2022] [Indexed: 11/23/2022] Open
Abstract
Several neurodegenerative diseases are characterized by the accumulation of aggregated misfolded proteins. These pathological agents have been suggested to propagate in the brain via mechanisms similar to that observed for the prion protein, where a misfolded variant is transferred from an affected brain region to a healthy one, thereby inducing the misfolding and/or aggregation of correctly folded copies. This process has been characterized for several proteins, such as α-synuclein, tau, amyloid beta (Aβ) and less extensively for huntingtin and TDP-43. α-synuclein, tau, TDP-43 and huntingtin are intracellular proteins, and their aggregates are located in the cytosol or nucleus of neurons. They have been shown to spread between cells and this event occurs, at least partially, via secretion of these protein aggregates in the extracellular space followed by re-uptake. Conversely, Aβ aggregates are found mainly extracellularly, and their spreading occurs in the extracellular space between brain regions. Due to the inherent nature of their spreading modalities, these proteins are exposed to components of the extracellular matrix (ECM), including glycans, proteases and core matrix proteins. These ECM components can interact with or process pathological misfolded proteins, potentially changing their properties and thus regulating their spreading capabilities. Here, we present an overview of the documented roles of ECM components in the spreading of pathological protein aggregates in neurodegenerative diseases with the objective of identifying the current gaps in knowledge and stimulating further research in the field. This could potentially lead to the identification of druggable targets to slow down the spreading and/or progression of these pathologies.
Collapse
Affiliation(s)
- Edoardo Moretto
- Institute of Neuroscience, National Research Council, CNR, Milan, Italy
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- *Correspondence: Edoardo Moretto,
| | - Skye Stuart
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
| | - Sunaina Surana
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
| | - Jose Norberto S. Vargas
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
| | - Giampietro Schiavo
- UK Dementia Research Institute, University College London, London, United Kingdom
- Department of Neuromuscular Diseases, Queen Square Institute of Neurology, University College London, London, United Kingdom
- UCL Queen Square Motor Neuron Disease Centre, University College London, London, United Kingdom
- Giampietro Schiavo,
| |
Collapse
|
24
|
Wankhede NL, Kale MB, Upaganlawar AB, Taksande BG, Umekar MJ, Behl T, Abdellatif AAH, Bhaskaran PM, Dachani SR, Sehgal A, Singh S, Sharma N, Makeen HA, Albratty M, Dailah HG, Bhatia S, Al-Harrasi A, Bungau S. Involvement of molecular chaperone in protein-misfolding brain diseases. Biomed Pharmacother 2022; 147:112647. [PMID: 35149361 DOI: 10.1016/j.biopha.2022.112647] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2021] [Revised: 01/03/2022] [Accepted: 01/12/2022] [Indexed: 12/19/2022] Open
Abstract
Protein misfolding causes aggregation and build-up in a variety of brain diseases. There are numeral molecules that are linked with the protein homeostasis mechanism. Molecular chaperones are one of such molecules that are responsible for protection against protein misfolded and aggregation-induced neurotoxicity. Many studies have explored the participation of molecular chaperones in Parkinson's disease, Alzheimer's disease, Amyotrophic lateral sclerosis, and Huntington's diseases. In this review, we highlighted the constructive role of molecular chaperones in neurological diseases characterized by protein misfolding and aggregation and their capability to control aberrant protein interactions at an early stage thus successfully suppressing pathogenic cascades. A comprehensive understanding of the protein misfolding associated with brain diseases and the molecular basis of involvement of chaperone against aggregation-induced cellular stress might lead to the progress of new therapeutic intrusion-related to protein misfolding and aggregation.
Collapse
Affiliation(s)
- Nitu L Wankhede
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra, India
| | - Mayur B Kale
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra, India
| | - Aman B Upaganlawar
- SNJB's Shriman Sureshdada Jain College of Pharmacy, Neminagar, Chandwad, Nasik, Maharashta, India
| | - Brijesh G Taksande
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra, India
| | - Milind J Umekar
- Smt. Kishoritai Bhoyar College of Pharmacy, Kamptee, Nagpur, Maharashtra, India
| | - Tapan Behl
- Chitkara College of Pharmacy, Chitkara University, Punjab, India.
| | - Ahmed A H Abdellatif
- Department of Pharmaceutics, College of Pharmacy, Qassim University, Buraydah, Saudi Arabia; Department of Pharmaceutics and Industrial Pharmacy, Faculty of Pharmacy, Al-Azhar University, Assiut, Egypt
| | | | - Sudarshan Reddy Dachani
- Department of Pharmacy Practice & Pharmacology, College of Pharmacy, Shaqra University (Al-Dawadmi Campus), Al-Dawadmi, Saudi Arabia
| | - Aayush Sehgal
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Sukhbir Singh
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Neelam Sharma
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Hafiz A Makeen
- Pharmacy Practice Research Unit, Clinical Pharmacy Department, College of Pharmacy, Jazan university, Jazan, Saudi Arabia
| | - Mohammed Albratty
- Department of Pharmaceutical Chemistry, College of Pharmacy, Jazan University, Jazan, Saudi Arabia
| | - Hamed Ghaleb Dailah
- Research and Scientific Studies Unit, College of Nursing, Jazan University, Jazan, Saudi Arabia
| | - Saurabh Bhatia
- Natural & Medical Sciences Research Center, University of Nizwa, Nizwa, Oman; School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Ahmed Al-Harrasi
- School of Health Science, University of Petroleum and Energy Studies, Dehradun, Uttarakhand, India
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania.
| |
Collapse
|
25
|
Lant JT, Kiri R, Duennwald ML, O'Donoghue P. Formation and persistence of polyglutamine aggregates in mistranslating cells. Nucleic Acids Res 2021; 49:11883-11899. [PMID: 34718744 PMCID: PMC8599886 DOI: 10.1093/nar/gkab898] [Citation(s) in RCA: 21] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/11/2021] [Revised: 09/03/2021] [Accepted: 09/20/2021] [Indexed: 12/17/2022] Open
Abstract
In neurodegenerative diseases, including pathologies with well-known causative alleles, genetic factors that modify severity or age of onset are not entirely understood. We recently documented the unexpected prevalence of transfer RNA (tRNA) mutants in the human population, including variants that cause amino acid mis-incorporation. We hypothesized that a mistranslating tRNA will exacerbate toxicity and modify the molecular pathology of Huntington's disease-causing alleles. We characterized a tRNAPro mutant that mistranslates proline codons with alanine, and tRNASer mutants, including a tRNASerAGA G35A variant with a phenylalanine anticodon (tRNASerAAA) found in ∼2% of the population. The tRNAPro mutant caused synthetic toxicity with a deleterious huntingtin poly-glutamine (polyQ) allele in neuronal cells. The tRNASerAAA variant showed synthetic toxicity with proteasome inhibition but did not enhance toxicity of the huntingtin allele. Cells mistranslating phenylalanine or proline codons with serine had significantly reduced rates of protein synthesis. Mistranslating cells were slow but effective in forming insoluble polyQ aggregates, defective in protein and aggregate degradation, and resistant to the neuroprotective integrated stress response inhibitor (ISRIB). Our findings identify mistranslating tRNA variants as genetic factors that slow protein aggregation kinetics, inhibit aggregate clearance, and increase drug resistance in cellular models of neurodegenerative disease.
Collapse
Affiliation(s)
- Jeremy T Lant
- Department of Biochemistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Rashmi Kiri
- Department of Biochemistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Martin L Duennwald
- Department of Anatomy & Cell Biology, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| | - Patrick O'Donoghue
- Department of Biochemistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada.,Department of Chemistry, The University of Western Ontario, London, Ontario N6A 5C1, Canada
| |
Collapse
|
26
|
Jarosińska OD, Rüdiger SGD. Molecular Strategies to Target Protein Aggregation in Huntington's Disease. Front Mol Biosci 2021; 8:769184. [PMID: 34869596 PMCID: PMC8636123 DOI: 10.3389/fmolb.2021.769184] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Accepted: 10/18/2021] [Indexed: 11/30/2022] Open
Abstract
Huntington's disease (HD) is a neurodegenerative disorder caused by the aggregation of the mutant huntingtin (mHTT) protein in nerve cells. mHTT self-aggregates to form soluble oligomers and insoluble fibrils, which interfere in a number of key cellular functions. This leads to cell quiescence and ultimately cell death. There are currently still no treatments available for HD, but approaches targeting the HTT levels offer systematic, mechanism-driven routes towards curing HD and other neurodegenerative diseases. This review summarizes the current state of knowledge of the mRNA targeting approaches such as antisense oligonucleotides and RNAi system; and the novel methods targeting mHTT and aggregates for degradation via the ubiquitin proteasome or the autophagy-lysosomal systems. These methods include the proteolysis-targeting chimera, Trim-Away, autophagosome-tethering compound, autophagy-targeting chimera, lysosome-targeting chimera and approach targeting mHTT for chaperone-mediated autophagy. These molecular strategies provide a knowledge-based approach to target HD and other neurodegenerative diseases at the origin.
Collapse
Affiliation(s)
- Olga D. Jarosińska
- Cellular Protein Chemistry, Bijvoet Centre for Biomolecular Research, Utrecht University, Utrecht, Netherlands
- Science for Life, Utrecht University, Utrecht, Netherlands
| | - Stefan G. D. Rüdiger
- Cellular Protein Chemistry, Bijvoet Centre for Biomolecular Research, Utrecht University, Utrecht, Netherlands
- Science for Life, Utrecht University, Utrecht, Netherlands
| |
Collapse
|
27
|
N-alpha-acetylation of Huntingtin protein increases its propensity to aggregate. J Biol Chem 2021; 297:101363. [PMID: 34732320 PMCID: PMC8640455 DOI: 10.1016/j.jbc.2021.101363] [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: 09/07/2021] [Revised: 10/23/2021] [Accepted: 10/25/2021] [Indexed: 11/22/2022] Open
Abstract
Huntington’s disease (HD) is a neurodegenerative disorder caused by a poly-CAG expansion in the first exon of the HTT gene, resulting in an extended poly-glutamine tract in the N-terminal domain of the Huntingtin (Htt) protein product. Proteolytic fragments of the poly-glutamine–containing N-terminal domain form intranuclear aggregates that are correlated with HD. Post-translational modification of Htt has been shown to alter its function and aggregation properties. However, the effect of N-terminal Htt acetylation has not yet been considered. Here, we developed a bacterial system to produce unmodified or N-terminally acetylated and aggregation-inducible Htt protein. We used this system together with biochemical, biophysical, and imaging studies to confirm that the Htt N-terminus is an in vitro substrate for the NatA N-terminal acetyltransferase and show that N-terminal acetylation promotes aggregation. These studies represent the first link between N-terminal acetylation and the promotion of a neurodegenerative disease and implicates NatA-mediated Htt acetylation as a new potential therapeutic target in HD.
Collapse
|
28
|
Vieweg S, Mahul-Mellier AL, Ruggeri FS, Riguet N, DeGuire SM, Chiki A, Cendrowska U, Dietler G, Lashuel HA. The Nt17 Domain and its Helical Conformation Regulate the Aggregation, Cellular Properties and Neurotoxicity of Mutant Huntingtin Exon 1. J Mol Biol 2021; 433:167222. [PMID: 34492254 DOI: 10.1016/j.jmb.2021.167222] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2021] [Revised: 08/25/2021] [Accepted: 08/26/2021] [Indexed: 11/29/2022]
Abstract
Converging evidence points to the N-terminal domain comprising the first 17 amino acids of the Huntingtin protein (Nt17) as a key regulator of its aggregation, cellular properties and toxicity. In this study, we further investigated the interplay between Nt17 and the polyQ domain repeat length in regulating the aggregation and inclusion formation of exon 1 of the Huntingtin protein (Httex1). In addition, we investigated the effect of removing Nt17 or modulating its local structure on the membrane interactions, neuronal uptake, and toxicity of monomeric or fibrillar Httex1. Our results show that the polyQ and Nt17 domains synergistically modulate the aggregation propensity of Httex1 and that the Nt17 domain plays important roles in shaping the surface properties of mutant Httex1 fibrils and regulating their poly-Q-dependent growth, lateral association and neuronal uptake. Removal of Nt17 or disruption of its transient helical conformations slowed the aggregation of monomeric Httex1 in vitro, reduced inclusion formation in cells, enhanced the neuronal uptake and nuclear accumulation of monomeric Httex1 proteins, and was sufficient to prevent cell death induced by Httex1 72Q overexpression. Finally, we demonstrate that the uptake of Httex1 fibrils into primary neurons and the resulting toxicity are strongly influenced by mutations and phosphorylation events that influence the local helical propensity of Nt17. Altogether, our results demonstrate that the Nt17 domain serves as one of the key master regulators of Htt aggregation, internalization, and toxicity and represents an attractive target for inhibiting Htt aggregate formation, inclusion formation, and neuronal toxicity.
Collapse
Affiliation(s)
- Sophie Vieweg
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Anne-Laure Mahul-Mellier
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Francesco S Ruggeri
- Laboratory of the Physics of Living Matter, EPFL, 1015 Lausanne, Switzerland
| | - Nathan Riguet
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Sean M DeGuire
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Anass Chiki
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland
| | - Urszula Cendrowska
- Laboratory of the Physics of Living Matter, EPFL, 1015 Lausanne, Switzerland
| | - Giovanni Dietler
- Laboratory of the Physics of Living Matter, EPFL, 1015 Lausanne, Switzerland
| | - Hilal A Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, Ecole Polytechnique Fédérale de Lausanne (EPFL), 1015 Lausanne, Switzerland.
| |
Collapse
|
29
|
Chongtham A, Isas JM, Pandey NK, Rawat A, Yoo JH, Mastro T, Kennedy MB, Langen R, Khoshnan A. Amplification of neurotoxic HTTex1 assemblies in human neurons. Neurobiol Dis 2021; 159:105517. [PMID: 34563643 DOI: 10.1016/j.nbd.2021.105517] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/20/2021] [Revised: 08/24/2021] [Accepted: 09/21/2021] [Indexed: 11/25/2022] Open
Abstract
Huntington's disease (HD) is a genetically inherited neurodegenerative disorder caused by expansion of a polyglutamine (polyQ) repeat in the exon-1 of huntingtin protein (HTT). The expanded polyQ enhances the amyloidogenic propensity of HTT exon 1 (HTTex1), which forms a heterogeneous mixture of assemblies with a broad neurotoxicity spectrum. While predominantly intracellular, monomeric and aggregated mutant HTT species are also present in the cerebrospinal fluids of HD patients, however, their biological properties are not well understood. To explore the role of extracellular mutant HTT in aggregation and toxicity, we investigated the uptake and amplification of recombinant HTTex1 assemblies in cell culture models. We find that small HTTex1 fibrils preferentially enter human neurons and trigger the amplification of neurotoxic assemblies; astrocytes or epithelial cells are not permissive. The amplification of HTTex1 in neurons depletes endogenous HTT protein with non-pathogenic polyQ repeat, activates apoptotic caspase-3 pathway and induces nuclear fragmentation. Using a panel of novel monoclonal antibodies and genetic mutation, we identified epitopes within the N-terminal 17 amino acids and proline-rich domain of HTTex1 to be critical in neural uptake and amplification. Synaptosome preparations from the brain homogenates of HD mice also contain mutant HTT species, which enter neurons and behave similar to small recombinant HTTex1 fibrils. These studies suggest that amyloidogenic extracellular mutant HTTex1 assemblies may preferentially enter neurons, propagate and promote neurodegeneration.
Collapse
Affiliation(s)
| | - J Mario Isas
- Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, CA 90089, USA
| | - Nitin K Pandey
- Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, CA 90089, USA
| | - Anoop Rawat
- Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, CA 90089, USA
| | - Jung Hyun Yoo
- Biology and Bioengineering, Caltech, Pasadena, CA 91125, USA
| | - Tara Mastro
- Biology and Bioengineering, Caltech, Pasadena, CA 91125, USA
| | - Mary B Kennedy
- Biology and Bioengineering, Caltech, Pasadena, CA 91125, USA
| | - Ralf Langen
- Zilkha Neurogenetic Institute, Keck School of Medicine of USC, Los Angeles, CA 90089, USA
| | - Ali Khoshnan
- Biology and Bioengineering, Caltech, Pasadena, CA 91125, USA.
| |
Collapse
|
30
|
Gerson JE, Safren N, Fischer S, Patel R, Crowley EV, Welday JP, Windle AK, Barmada S, Paulson HL, Sharkey LM. Ubiquilin-2 differentially regulates polyglutamine disease proteins. Hum Mol Genet 2021; 29:2596-2610. [PMID: 32681165 DOI: 10.1093/hmg/ddaa152] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 07/07/2020] [Accepted: 07/13/2020] [Indexed: 12/12/2022] Open
Abstract
Divergent protein context helps explain why polyglutamine expansion diseases differ clinically and pathologically. This heterogeneity may also extend to how polyglutamine disease proteins are handled by cellular pathways of proteostasis. Studies suggest, for example, that the ubiquitin-proteasome shuttle protein Ubiquilin-2 (UBQLN2) selectively interacts with specific polyglutamine disease proteins. Here we employ cellular models, primary neurons and mouse models to investigate the potential differential regulation by UBQLN2 of two polyglutamine disease proteins, huntingtin (HTT) and ataxin-3 (ATXN3). In cells, overexpressed UBQLN2 selectively lowered levels of full-length pathogenic HTT but not of HTT exon 1 fragment or full-length ATXN3. Consistent with these results, UBQLN2 specifically reduced accumulation of aggregated mutant HTT but not mutant ATXN3 in mouse models of Huntington's disease (HD) and spinocerebellar ataxia type 3 (SCA3), respectively. Normally a cytoplasmic protein, UBQLN2 translocated to the nuclei of neurons in HD mice but not in SCA3 mice. Remarkably, instead of reducing the accumulation of nuclear mutant ATXN3, UBQLN2 induced an accumulation of cytoplasmic ATXN3 aggregates in neurons of SCA3 mice. Together these results reveal a selective action of UBQLN2 toward polyglutamine disease proteins, indicating that polyglutamine expansion alone is insufficient to promote UBQLN2-mediated clearance of this class of disease proteins. Additional factors, including nuclear translocation of UBQLN2, may facilitate its action to clear intranuclear, aggregated disease proteins like HTT.
Collapse
Affiliation(s)
- Julia E Gerson
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Nathaniel Safren
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Svetlana Fischer
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Ronak Patel
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Emily V Crowley
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Jacqueline P Welday
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Alexandra K Windle
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Sami Barmada
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Henry L Paulson
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| | - Lisa M Sharkey
- Department of Neurology, University of Michigan, Ann Arbor, MI 48109-2200, USA
| |
Collapse
|
31
|
Chongtham A, Bornemann DJ, Barbaro BA, Lukacsovich T, Agrawal N, Syed A, Worthge S, Purcell J, Burke J, Chin TM, Marsh JL. Effects of flanking sequences and cellular context on subcellular behavior and pathology of mutant HTT. Hum Mol Genet 2021; 29:674-688. [PMID: 31943010 DOI: 10.1093/hmg/ddaa001] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2019] [Revised: 12/06/2019] [Accepted: 01/07/2020] [Indexed: 12/11/2022] Open
Abstract
Huntington's disease (HD) is caused by an expansion of a poly glutamine (polyQ) stretch in the huntingtin protein (HTT) that is necessary to cause pathology and formation of HTT aggregates. Here we ask whether expanded polyQ is sufficient to cause pathology and aggregate formation. By addressing the sufficiency question, one can identify cellular processes and structural parameters that influence HD pathology and HTT subcellular behavior (i.e. aggregation state and subcellular location). Using Drosophila, we compare the effects of expressing mutant full-length human HTT (fl-mHTT) to the effects of mutant human HTTexon1 and to two commonly used synthetic fragments, HTT171 and shortstop (HTT118). Expanded polyQ alone is not sufficient to cause inclusion formation since full-length HTT and HTTex1 with expanded polyQ are both toxic although full-length HTT remains diffuse while HTTex1 forms inclusions. Further, inclusions are not sufficient to cause pathology since HTT171-120Q forms inclusions but is benign and co-expression of HTT171-120Q with non-aggregating pathogenic fl-mHTT recruits fl-mHTT to aggregates and rescues its pathogenicity. Additionally, the influence of sequences outside the expanded polyQ domain is revealed by finding that small modifications to the HTT118 or HTT171 fragments can dramatically alter their subcellular behavior and pathogenicity. Finally, mutant HTT subcellular behavior is strongly modified by different cell and tissue environments (e.g. fl-mHTT appears as diffuse nuclear in one tissue and diffuse cytoplasmic in another but toxic in both). These observations underscore the importance of cellular and structural context for the interpretation and comparison of experiments using different fragments and tissues to report the effects of expanded polyQ.
Collapse
Affiliation(s)
- Anjalika Chongtham
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, 92697-2300, USA.,Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125
| | - Douglas J Bornemann
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, 92697-2300, USA
| | - Brett A Barbaro
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, 92697-2300, USA
| | - Tamas Lukacsovich
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, 92697-2300, USA
| | - Namita Agrawal
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, 92697-2300, USA
| | - Adeela Syed
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, 92697-2300, USA
| | - Shane Worthge
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, 92697-2300, USA
| | - Judith Purcell
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, 92697-2300, USA
| | - John Burke
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, 92697-2300, USA
| | - Theodore M Chin
- Division of Biology and Biological Engineering, California Institute of Technology, Pasadena, CA, 91125
| | - J Lawrence Marsh
- Department of Developmental and Cell Biology, University of California Irvine, Irvine, CA, 92697-2300, USA
| |
Collapse
|
32
|
Onur TS, Laitman A, Zhao H, Keyho R, Kim H, Wang J, Mair M, Wang H, Li L, Perez A, de Haro M, Wan YW, Allen G, Lu B, Al-Ramahi I, Liu Z, Botas J. Downregulation of glial genes involved in synaptic function mitigates Huntington's disease pathogenesis. eLife 2021; 10:64564. [PMID: 33871358 PMCID: PMC8149125 DOI: 10.7554/elife.64564] [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: 11/03/2020] [Accepted: 04/19/2021] [Indexed: 01/01/2023] Open
Abstract
Most research on neurodegenerative diseases has focused on neurons, yet glia help form and maintain the synapses whose loss is so prominent in these conditions. To investigate the contributions of glia to Huntington's disease (HD), we profiled the gene expression alterations of Drosophila expressing human mutant Huntingtin (mHTT) in either glia or neurons and compared these changes to what is observed in HD human and HD mice striata. A large portion of conserved genes are concordantly dysregulated across the three species; we tested these genes in a high-throughput behavioral assay and found that downregulation of genes involved in synapse assembly mitigated pathogenesis and behavioral deficits. To our surprise, reducing dNRXN3 function in glia was sufficient to improve the phenotype of flies expressing mHTT in neurons, suggesting that mHTT's toxic effects in glia ramify throughout the brain. This supports a model in which dampening synaptic function is protective because it attenuates the excitotoxicity that characterizes HD. When a neuron dies, through injury or disease, the body loses all communication that passes through it. The brain compensates by rerouting the flow of information through other neurons in the network. Eventually, if the loss of neurons becomes too great, compensation becomes impossible. This process happens in Alzheimer's, Parkinson's, and Huntington's disease. In the case of Huntington's disease, the cause is mutation to a single gene known as huntingtin. The mutation is present in every cell in the body but causes particular damage to parts of the brain involved in mood, thinking and movement. Neurons and other cells respond to mutations in the huntingtin gene by turning the activities of other genes up or down, but it is not clear whether all of these changes contribute to the damage seen in Huntington's disease. In fact, it is possible that some of the changes are a result of the brain trying to protect itself. So far, most research on this subject has focused on neurons because the huntingtin gene plays a role in maintaining healthy neuronal connections. But, given that all cells carry the mutated gene, it is likely that other cells are also involved. The glia are a diverse group of cells that support the brain, providing care and sustenance to neurons. These cells have a known role in maintaining the connections between neurons and may also have play a role in either causing or correcting the damage seen in Huntington's disease. The aim of Onur et al. was to find out which genes are affected by having a mutant huntingtin gene in neurons or glia, and whether severity of Huntington’s disease improved or worsened when the activity of these genes changed. First, Onur et al. identified genes affected by mutant huntingtin by comparing healthy human brains to the brains of people with Huntington's disease. Repeating the same comparison in mice and fruit flies identified genes affected in the same way across all three species, revealing that, in Huntington's disease, the brain dials down glial cell genes involved in maintaining neuronal connections. To find out how these changes in gene activity affect disease severity and progression, Onur et al. manipulated the activity of each of the genes they had identified in fruit flies that carried mutant versions of huntingtin either in neurons, in glial cells or in both cell types. They then filmed the flies to see the effects of the manipulation on movement behaviors, which are affected by Huntington’s disease. This revealed that purposely lowering the activity of the glial genes involved in maintaining connections between neurons improved the symptoms of the disease, but only in flies who had mutant huntingtin in their glial cells. This indicates that the drop in activity of these genes observed in Huntington’s disease is the brain trying to protect itself. This work suggests that it is important to include glial cells in studies of neurological disorders. It also highlights the fact that changes in gene expression as a result of a disease are not always bad. Many alterations are compensatory, and try to either make up for or protect cells affected by the disease. Therefore, it may be important to consider whether drugs designed to treat a condition by changing levels of gene activity might undo some of the body's natural protection. Working out which changes drive disease and which changes are protective will be essential for designing effective treatments.
Collapse
Affiliation(s)
- Tarik Seref Onur
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States.,Genetics & Genomics Graduate Program, Baylor College of Medicine, Houston, United States
| | - Andrew Laitman
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States.,Quantitative & Computational Biosciences, Baylor College of Medicine, Houston, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, United States
| | - He Zhao
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Ryan Keyho
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Hyemin Kim
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Jennifer Wang
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Megan Mair
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States.,Genetics & Genomics Graduate Program, Baylor College of Medicine, Houston, United States
| | - Huilan Wang
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Lifang Li
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Alma Perez
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Maria de Haro
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Ying-Wooi Wan
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Genevera Allen
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States.,Departments of Electrical & Computer Engineering, Statistics and Computer Science, Rice University, Houston, United States
| | - Boxun Lu
- State Key Laboratory of Medical Neurobiology and MOE Frontiers Center for Brain Science, Fudan University, Shanghai, China
| | - Ismael Al-Ramahi
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States
| | - Zhandong Liu
- Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States.,Quantitative & Computational Biosciences, Baylor College of Medicine, Houston, United States.,Department of Pediatrics, Baylor College of Medicine, Houston, United States
| | - Juan Botas
- Department of Molecular and Human Genetics, Baylor College of Medicine, Houston, United States.,Jan and Dan Duncan Neurological Research Institute at Texas Children's Hospital, Houston, United States.,Genetics & Genomics Graduate Program, Baylor College of Medicine, Houston, United States.,Quantitative & Computational Biosciences, Baylor College of Medicine, Houston, United States
| |
Collapse
|
33
|
Zhou Y, Peskett TR, Landles C, Warner JB, Sathasivam K, Smith EJ, Chen S, Wetzel R, Lashuel HA, Bates GP, Saibil HR. Correlative light and electron microscopy suggests that mutant huntingtin dysregulates the endolysosomal pathway in presymptomatic Huntington's disease. Acta Neuropathol Commun 2021; 9:70. [PMID: 33853668 PMCID: PMC8048291 DOI: 10.1186/s40478-021-01172-z] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/10/2021] [Accepted: 03/28/2021] [Indexed: 12/18/2022] Open
Abstract
Huntington's disease (HD) is a late onset, inherited neurodegenerative disorder for which early pathogenic events remain poorly understood. Here we show that mutant exon 1 HTT proteins are recruited to a subset of cytoplasmic aggregates in the cell bodies of neurons in brain sections from presymptomatic HD, but not wild-type, mice. This occurred in a disease stage and polyglutamine-length dependent manner. We successfully adapted a high-resolution correlative light and electron microscopy methodology, originally developed for mammalian and yeast cells, to allow us to correlate light microscopy and electron microscopy images on the same brain section within an accuracy of 100 nm. Using this approach, we identified these recruitment sites as single membrane bound, vesicle-rich endolysosomal organelles, specifically as (1) multivesicular bodies (MVBs), or amphisomes and (2) autolysosomes or residual bodies. The organelles were often found in close-proximity to phagophore-like structures. Immunogold labeling localized mutant HTT to non-fibrillar, electron lucent structures within the lumen of these organelles. In presymptomatic HD, the recruitment organelles were predominantly MVBs/amphisomes, whereas in late-stage HD, there were more autolysosomes or residual bodies. Electron tomograms indicated the fusion of small vesicles with the vacuole within the lumen, suggesting that MVBs develop into residual bodies. We found that markers of MVB-related exocytosis were depleted in presymptomatic mice and throughout the disease course. This suggests that endolysosomal homeostasis has moved away from exocytosis toward lysosome fusion and degradation, in response to the need to clear the chronically aggregating mutant HTT protein, and that this occurs at an early stage in HD pathogenesis.
Collapse
Affiliation(s)
- Ya Zhou
- Huntington’s Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, University College London, London, UK
| | - Thomas R. Peskett
- Institute of Structural and Molecular Biology, Birkbeck College, London, WC1E 7HX UK
- Present Address: Department of Biology, Institute of Biochemistry, ETH Zurich, Otto-Stern-Weg 3, 8093 Zurich, Switzerland
| | - Christian Landles
- Huntington’s Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, University College London, London, UK
| | - John B. Warner
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Kirupa Sathasivam
- Huntington’s Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, University College London, London, UK
| | - Edward J. Smith
- Huntington’s Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, University College London, London, UK
| | - Shu Chen
- Institute of Structural and Molecular Biology, Birkbeck College, London, WC1E 7HX UK
| | - Ronald Wetzel
- Department of Structural Biology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15260 USA
| | - Hilal A. Lashuel
- Laboratory of Molecular and Chemical Biology of Neurodegeneration, Brain Mind Institute, École Polytechnique Fédérale de Lausanne (EPFL), Lausanne, Switzerland
| | - Gillian P. Bates
- Huntington’s Disease Centre, Department of Neurodegenerative Disease and UK Dementia Research Institute at UCL, Queen Square Institute of Neurology, University College London, London, UK
| | - Helen R. Saibil
- Institute of Structural and Molecular Biology, Birkbeck College, London, WC1E 7HX UK
| |
Collapse
|
34
|
Implications of the Orb2 Amyloid Structure in Huntington's Disease. Int J Mol Sci 2020; 21:ijms21186910. [PMID: 32967102 PMCID: PMC7555547 DOI: 10.3390/ijms21186910] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Revised: 09/11/2020] [Accepted: 09/16/2020] [Indexed: 02/06/2023] Open
Abstract
Huntington’s disease is a progressive, autosomal dominant, neurodegenerative disorder caused by an expanded CAG repeat in the huntingtin gene. As a result, the translated protein, huntingtin, contains an abnormally long polyglutamine stretch that makes it prone to misfold and aggregating. Aggregation of huntingtin is believed to be the cause of Huntington’s disease. However, understanding on how, and why, huntingtin aggregates are deleterious has been hampered by lack of enough relevant structural data. In this review, we discuss our recent findings on a glutamine-based functional amyloid isolated from Drosophila brain and how this information provides plausible structural insight on the structure of huntingtin deposits in the brain.
Collapse
|
35
|
Bartl S, Oueslati A, Southwell AL, Siddu A, Parth M, David LS, Maxan A, Salhat N, Burkert M, Mairhofer A, Friedrich T, Pankevych H, Balazs K, Staffler G, Hayden MR, Cicchetti F, Smrzka OW. Inhibiting cellular uptake of mutant huntingtin using a monoclonal antibody: Implications for the treatment of Huntington's disease. Neurobiol Dis 2020; 141:104943. [PMID: 32407769 DOI: 10.1016/j.nbd.2020.104943] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Revised: 04/08/2020] [Accepted: 05/06/2020] [Indexed: 12/24/2022] Open
Abstract
Huntington's disease (HD) is caused by a highly polymorphic CAG trinucleotide expansion in the gene encoding for the huntingtin protein (HTT). The resulting mutant huntingtin protein (mutHTT) is ubiquitously expressed but also exhibits the ability to propagate from cell-to-cell to disseminate pathology; a property which may serve as a new therapeutic focus. Accordingly, we set out to develop a monoclonal antibody (mAB) targeting a particularly exposed region close to the aa586 caspase-6 cleavage site of the HTT protein. This monoclonal antibody, designated C6-17, effectively binds mutHTT and is able to deplete the protein from cell culture supernatants. Using cell-based assays, we demonstrate that extracellular secretion of mutHTT into cell culture media and its subsequent uptake in recipient HeLa cells can be almost entirely blocked by mAB C6-17. Immunohistochemical stainings of post-mortem HD brain tissue confirmed the specificity of mAB C6-17 to human mutHTT aggregates. These findings demonstrate that mAB C6-17 not only successfully engages with its target, mutHTT, but also inhibits cell uptake suggesting that this antibody could interfere with the pathological processes of mutHTT spreading in vivo.
Collapse
Affiliation(s)
| | - Abid Oueslati
- Université Laval/Centre de recherche du CHU, Québec, Canada
| | | | - Alberto Siddu
- Université Laval/Centre de recherche du CHU, Québec, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | | - Oskar W Smrzka
- AFFiRiS AG, Vienna, Austria; Ablevia biotech GmbH, Vienna, Austria
| |
Collapse
|
36
|
Tang BL. Could metformin be therapeutically useful in Huntington's disease? Rev Neurosci 2020; 31:297-317. [PMID: 31751298 DOI: 10.1515/revneuro-2019-0072] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2019] [Accepted: 09/09/2019] [Indexed: 12/15/2022]
Abstract
Emerging evidence suggest that dimethylbiguanide (metformin), a first-line drug for type 2 diabetes mellitus, could be neuroprotective in a range of brain pathologies, which include neurodegenerative diseases and brain injury. However, there are also contraindications that associate metformin treatment with cognitive impairment as well as adverse outcomes in Alzheimer's disease and Parkinson's disease animal models. Recently, a beneficial effect of metformin in animal models of Huntington's disease (HD) has been strengthened by multiple reports. In this brief review, the findings associated with the effects of metformin in attenuating neurodegenerative diseases are discussed, focusing on HD-associated pathology and the potential underlying mechanisms highlighted by these studies. The mechanism of action of metformin is complex, and its therapeutic efficacy is therefore expected to be dependent on the disease context. The key metabolic pathways that are effectively affected by metformin, such as AMP-activated protein kinase activation, may be altered in the later decades of the human lifespan. In this regard, metformin may nonetheless be therapeutically useful for neurological diseases with early pathological onsets, such as HD.
Collapse
Affiliation(s)
- Bor Luen Tang
- Department of Biochemistry, Yong Loo Lin School of Medicine, National University Health System, Singapore 117596, Singapore.,NUS Graduate School for Integrative Sciences and Engineering, National University of Singapore, Medical Drive, Singapore 119077, Singapore
| |
Collapse
|
37
|
Assessing average somatic CAG repeat instability at the protein level. Sci Rep 2019; 9:19152. [PMID: 31844074 PMCID: PMC6915696 DOI: 10.1038/s41598-019-55202-x] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2019] [Accepted: 11/18/2019] [Indexed: 12/25/2022] Open
Abstract
Sandwich ELISA-based methods use Abs that target the expanded polyglutamine (polyQ) tract to quantify mutant huntingtin (mHTT). Using Meso Scale Discovery (MSD) assay, the mHTT signal detected with MW1 Ab correlated with polyQ length and doubled with a difference of only 7 glutamine residues between equivalent amounts of purified mHTTexon1 proteins. Similar polyQ length-dependent effects on MSD signals were confirmed using endogenous full length mHTT from brains of Huntington’s disease (HD) knock-in (KI) mice. We used this avidity bias to devise a method to assess average CAG repeat instability at the protein level in a mixed population of HTT proteins present in tissues. Signal detected for average polyQ length quantification at the protein level by our method exhibited a strong correlation with average CAG repeat length at the genomic DNA level determined by PCR method in striatal tissue homogenates from HdhQ140 KI mice and in human HD postmortem cortex. This work establishes that CAG repeat instability in mutant HTT is reflected at the protein level.
Collapse
|
38
|
Ekman FK, Ojala DS, Adil MM, Lopez PA, Schaffer DV, Gaj T. CRISPR-Cas9-Mediated Genome Editing Increases Lifespan and Improves Motor Deficits in a Huntington's Disease Mouse Model. MOLECULAR THERAPY. NUCLEIC ACIDS 2019; 17:829-839. [PMID: 31465962 PMCID: PMC6717077 DOI: 10.1016/j.omtn.2019.07.009] [Citation(s) in RCA: 101] [Impact Index Per Article: 16.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/12/2018] [Revised: 04/29/2019] [Accepted: 07/16/2019] [Indexed: 12/11/2022]
Abstract
Huntington's disease (HD) is a currently incurable and, ultimately, fatal neurodegenerative disorder caused by a CAG trinucleotide repeat expansion within exon 1 of the huntingtin (HTT) gene, which results in the production of a mutant protein that forms inclusions and selectively destroys neurons in the striatum and other adjacent structures. The RNA-guided Cas9 endonuclease from CRISPR-Cas9 systems is a versatile technology for inducing DNA double-strand breaks that can stimulate the introduction of frameshift-inducing mutations and permanently disable mutant gene function. Here, we show that the Cas9 nuclease from Staphylococcus aureus, a small Cas9 ortholog that can be packaged alongside a single guide RNA into a single adeno-associated virus (AAV) vector, can be used to disrupt the expression of the mutant HTT gene in the R6/2 mouse model of HD following its in vivo delivery to the striatum. Specifically, we found that CRISPR-Cas9-mediated disruption of the mutant HTT gene resulted in a ∼50% decrease in neuronal inclusions and significantly improved lifespan and certain motor deficits. These results thus illustrate the potential for CRISPR-Cas9 technology to treat HD and other autosomal dominant neurodegenerative disorders caused by a trinucleotide repeat expansion via in vivo genome editing.
Collapse
Affiliation(s)
- Freja K Ekman
- Department of Chemistry, University of California, Berkeley, Berkeley, CA, USA
| | - David S Ojala
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA
| | - Maroof M Adil
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA
| | - Paola A Lopez
- Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA
| | - David V Schaffer
- Department of Chemical and Biomolecular Engineering, University of California, Berkeley, Berkeley, CA, USA; Department of Bioengineering, University of California, Berkeley, Berkeley, CA, USA; Department of Molecular and Cell Biology, University of California, Berkeley, Berkeley, CA, USA; The Helen Wills Neuroscience Institute, University of California, Berkeley, Berkeley, CA, USA.
| | - Thomas Gaj
- Department of Bioengineering, University of Illinois, Urbana, IL, USA; Carl R. Woese Institute for Genomic Biology, University of Illinois, Urbana, IL, USA.
| |
Collapse
|
39
|
Sap KA, Guler AT, Bezstarosti K, Bury AE, Juenemann K, Demmers JA, Reits EA. Global Proteome and Ubiquitinome Changes in the Soluble and Insoluble Fractions of Q175 Huntington Mice Brains. Mol Cell Proteomics 2019; 18:1705-1720. [PMID: 31138642 PMCID: PMC6731087 DOI: 10.1074/mcp.ra119.001486] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2019] [Revised: 05/21/2019] [Indexed: 01/31/2023] Open
Abstract
Huntington's disease is caused by a polyglutamine repeat expansion in the huntingtin protein which affects the function and folding of the protein, and results in intracellular protein aggregates. Here, we examined whether this mutation leads to altered ubiquitination of huntingtin and other proteins in both soluble and insoluble fractions of brain lysates of the Q175 knock-in Huntington's disease mouse model and the Q20 wild-type mouse model. Ubiquitination sites are detected by identification of Gly-Gly (diGly) remnant motifs that remain on modified lysine residues after digestion. We identified K6, K9, K132, K804, and K837 as endogenous ubiquitination sites of soluble huntingtin, with wild-type huntingtin being mainly ubiquitinated at K132, K804, and K837. Mutant huntingtin protein levels were strongly reduced in the soluble fraction whereas K6 and K9 were mainly ubiquitinated. In the insoluble fraction increased levels of huntingtin K6 and K9 diGly sites were observed for mutant huntingtin as compared with wild type. Besides huntingtin, proteins with various roles, including membrane organization, transport, mRNA processing, gene transcription, translation, catabolic processes and oxidative phosphorylation, were differently expressed or ubiquitinated in wild-type and mutant huntingtin brain tissues. Correlating protein and diGly site fold changes in the soluble fraction revealed that diGly site abundances of most of the proteins were not related to protein fold changes, indicating that these proteins were differentially ubiquitinated in the Q175 mice. In contrast, both the fold change of the protein level and diGly site level were increased for several proteins in the insoluble fraction, including ubiquitin, ubiquilin-2, sequestosome-1/p62 and myo5a. Our data sheds light on putative novel proteins involved in different cellular processes as well as their ubiquitination status in Huntington's disease, which forms the basis for further mechanistic studies to understand the role of differential ubiquitination of huntingtin and ubiquitin-regulated processes in Huntington's disease.
Collapse
Affiliation(s)
- Karen A Sap
- ‡Department of Medical Biology, Amsterdam UMC, location AMC, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Arzu Tugce Guler
- ‡Department of Medical Biology, Amsterdam UMC, location AMC, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Karel Bezstarosti
- §Department of Biochemistry, Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Aleksandra E Bury
- ‡Department of Medical Biology, Amsterdam UMC, location AMC, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands
| | - Katrin Juenemann
- ¶Leibniz-Forschungsinstitut für Molekulare Pharmakologie im Forschungsverbund Berlin, Robert-Roessle-St. 10 13089 Berlin, Germany
| | - JeroenA A Demmers
- §Department of Biochemistry, Erasmus University Medical Center, Wytemaweg 80, 3015 CN Rotterdam, The Netherlands
| | - Eric A Reits
- ‡Department of Medical Biology, Amsterdam UMC, location AMC, Meibergdreef 15, 1105 AZ Amsterdam, The Netherlands.
| |
Collapse
|
40
|
Tao M, Pandey NK, Barnes R, Han S, Langen R. Structure of Membrane-Bound Huntingtin Exon 1 Reveals Membrane Interaction and Aggregation Mechanisms. Structure 2019; 27:1570-1580.e4. [PMID: 31466833 DOI: 10.1016/j.str.2019.08.003] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2019] [Revised: 07/23/2019] [Accepted: 08/09/2019] [Indexed: 02/08/2023]
Abstract
Huntington's disease is caused by a polyQ expansion in the first exon of huntingtin (Httex1). Membrane interaction of huntingtin is of physiological and pathological relevance. Using electron paramagnetic resonance and Overhauser dynamic nuclear polarization, we find that the N-terminal residues 3-13 of wild-type Httex1(Q25) form a membrane-bound, amphipathic α helix. This helix is positioned in the interfacial region, where it is sensitive to membrane curvature and electrostatic interactions with head-group charges. Residues 14-22, which contain the first five residues of the polyQ region, are in a transition region that remains in the interfacial region without taking up a stable, α-helical structure. The remaining C-terminal portion is solvent exposed. The phosphomimetic S13D/S16D mutations, which are known to protect from toxicity, inhibit membrane binding and attenuate membrane-mediated aggregation of mutant Httex1(Q46) due to electrostatic repulsion. Targeting the N-terminal membrane anchor using post-translational modifications or specific binders could be a potential means to reduce aggregation and toxicity in vivo.
Collapse
Affiliation(s)
- Meixin Tao
- Department of Neuroscience and Physiology, Department of Biochemistry and Molecular Medicine, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Nitin K Pandey
- Department of Neuroscience and Physiology, Department of Biochemistry and Molecular Medicine, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA
| | - Ryan Barnes
- Department of Chemistry and Biochemistry, Department of Chemical Engineering, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Songi Han
- Department of Chemistry and Biochemistry, Department of Chemical Engineering, University of California Santa Barbara, Santa Barbara, CA 93106, USA
| | - Ralf Langen
- Department of Neuroscience and Physiology, Department of Biochemistry and Molecular Medicine, Zilkha Neurogenetic Institute, Keck School of Medicine, University of Southern California, Los Angeles, CA 90033, USA.
| |
Collapse
|
41
|
Liu P, Smith BR, Huang ES, Mahesh A, Vonsattel JPG, Petersen AJ, Gomez-Pastor R, Ashe KH. A soluble truncated tau species related to cognitive dysfunction and caspase-2 is elevated in the brain of Huntington's disease patients. Acta Neuropathol Commun 2019; 7:111. [PMID: 31358058 PMCID: PMC6664763 DOI: 10.1186/s40478-019-0764-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2019] [Accepted: 07/01/2019] [Indexed: 11/18/2022] Open
Abstract
Huntington's disease (HD) is a progressive neurodegenerative disease. Involuntary movements, cognitive impairment and psychiatric disturbance are the major clinical manifestations, and gradual atrophy and selective neuronal loss in the striatum and cerebral cortex are the pathologic hallmarks. HD is caused by expanded CAG trinucleotide repeats at the N-terminus of IT15 that encodes the huntingtin (HTT) protein, though the molecular mechanisms through which the mutant HTT (mHTT) exerts toxic effects remain obscure. Members of the caspase family, including caspase-2 (Casp2), play an important role in HD pathogenesis. Genetic ablation of Casp2 ameliorates cognitive and motor deficits of HD mice, though the molecular targets of Casp2 are still unclear. It is well established that the microtubule-associated protein tau potentiates cognitive dysfunction in a variety of neurodegenerative disorders, including HD. Our recent study indicates that Casp2-catalyzed tau cleavage at aspartate 314 (tau 2N4R isoform numbering system) mediates synaptotoxicity, cognitive deficits and neurodegeneration in cellular and mouse models of frontotemporal dementia; further, levels of Δtau314, the soluble, N-terminal cleavage product, are elevated in individuals with mild cognitive impairment and Alzheimer's disease, compared with cognitively normal individuals. Here, we identified the presence of Δtau314 proteins in the striatum (caudate nucleus) and prefrontal cortex (Brodmann's area 8/9) of human subjects, and showed that in both structures, levels of Casp2 and Δtau314 proteins correlate well, and both proteins are higher in HD patients than non-HD individuals. Our findings advance our understanding of the contribution of Casp2-mediated Δtau314 production to HD pathogenesis.
Collapse
Affiliation(s)
- Peng Liu
- Departments of Neurology, University of Minnesota, Minneapolis, MN 55455 USA
- N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN 55455 USA
| | - Benjamin R. Smith
- Departments of Neurology, University of Minnesota, Minneapolis, MN 55455 USA
- N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN 55455 USA
| | - Eric S. Huang
- Departments of Neurology, University of Minnesota, Minneapolis, MN 55455 USA
- N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN 55455 USA
- Current address: University of Miami, Miller School of Medicine, Miami, FL 33136 USA
| | | | - Jean Paul G. Vonsattel
- Department of Pathology, Presbyterian Hospital and Columbia University, New York City, NY 10032 USA
| | - Ashley J. Petersen
- Division of Biostatistics, University of Minnesota, Minneapolis, MN 55455 USA
| | - Rocio Gomez-Pastor
- Departments of Neuroscience, University of Minnesota, Minneapolis, MN 55455 USA
| | - Karen H. Ashe
- Departments of Neurology, University of Minnesota, Minneapolis, MN 55455 USA
- Departments of Neuroscience, University of Minnesota, Minneapolis, MN 55455 USA
- N. Bud Grossman Center for Memory Research and Care, University of Minnesota, Minneapolis, MN 55455 USA
- Geriatric Research, Education, and Clinical Centers, Veterans Affairs Medical Center, Minneapolis, MN 55417 USA
| |
Collapse
|
42
|
Sebastián-Serrano Á, Simón-García A, Belmonte-Alfaro A, Pose-Utrilla J, Santos-Galindo M, Del Puerto A, García-Guerra L, Hernández IH, Schiavo G, Campanero MR, Lucas JJ, Iglesias T. Differential regulation of Kidins220 isoforms in Huntington's disease. Brain Pathol 2019; 30:120-136. [PMID: 31264746 DOI: 10.1111/bpa.12761] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2019] [Accepted: 06/25/2019] [Indexed: 02/06/2023] Open
Abstract
Huntington's disease (HD) is an inherited progressive neurodegenerative disease characterized by brain atrophy particularly in the striatum that produces motor impairment, and cognitive and psychiatric disturbances. Multiple pathogenic mechanisms have been proposed including dysfunctions in neurotrophic support and calpain-overactivation, among others. Kinase D-interacting substrate of 220 kDa (Kidins220), also known as ankyrin repeat-rich membrane spanning (ARMS), is an essential mediator of neurotrophin signaling. In adult brain, Kidins220 presents two main isoforms that differ in their carboxy-terminal length and critical protein-protein interaction domains. These variants are generated through alternative terminal exon splicing of the conventional exon 32 (Kidins220-C32) and the recently identified exon 33 (Kidins220-C33). The lack of domains encoded by exon 32 involved in key neuronal functions, including those controlling neurotrophin pathways, pointed to Kidins220-C33 as a form detrimental for neurons. However, the functional role of Kidins220-C33 in neurodegeneration or other pathologies, including HD, has not been explored. In the present work, we discover an unexpected selective downregulation of Kidins220-C33, in the striatum of HD patients, as well as in the R6/1 HD mouse model starting at early symptomatic stages. These changes are C33-specific as Kidins220-C32 variant remains unchanged. We also find the early decrease in Kidins220-C33 levels takes place in neurons, suggesting an unanticipated neuroprotective role for this isoform. Finally, using ex vivo assays and primary neurons, we demonstrate that Kidins220-C33 is downregulated by mechanisms that depend on the activation of the protease calpain. Altogether, these results strongly suggest that calpain-mediated Kidins220-C33 proteolysis modulates onset and/or progression of HD.
Collapse
Affiliation(s)
- Álvaro Sebastián-Serrano
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Ana Simón-García
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Alicia Belmonte-Alfaro
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Julia Pose-Utrilla
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - María Santos-Galindo
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain
| | - Ana Del Puerto
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Lucía García-Guerra
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| | - Ivó H Hernández
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain.,Facultad de Ciencias, Departamento de Biología (Unidad Docente Fisiología Animal), Universidad Autónoma de Madrid, Madrid, Spain
| | - Giampietro Schiavo
- Department of Neuromuscular Disorders, UCL Institute of Neurology, University College London, London, UK
| | - Miguel R Campanero
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red en Enfermedades Cardiovasculares (CIBERCV), Madrid, Spain
| | - José J Lucas
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain.,Centro de Biología Molecular "Severo Ochoa" (CSIC-UAM), Madrid, Spain
| | - Teresa Iglesias
- Instituto de Investigaciones Biomédicas "Alberto Sols", Consejo Superior de Investigaciones Científicas-Universidad Autónoma de Madrid (CSIC-UAM), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto de Salud Carlos III, Madrid, Spain
| |
Collapse
|
43
|
Peng W, Minakaki G, Nguyen M, Krainc D. Preserving Lysosomal Function in the Aging Brain: Insights from Neurodegeneration. Neurotherapeutics 2019; 16:611-634. [PMID: 31183763 PMCID: PMC6694346 DOI: 10.1007/s13311-019-00742-3] [Citation(s) in RCA: 59] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Lysosomes are acidic, membrane-bound organelles that serve as the primary catabolic compartment of the cell. They are crucial to a variety of cellular processes from nutrient storage to autophagy. Given the diversity of lysosomal functions, it is unsurprising that lysosomes are also emerging as important players in aging. Lysosomal dysfunction is implicated in several aging-related neurodegenerative diseases including Alzheimer's, Parkinson's, amyotrophic lateral sclerosis/frontotemporal dementia, and Huntington's. Although the precise role of lysosomes in the aging brain is not well-elucidated, some insight into their function has been gained from our understanding of the pathophysiology of age-dependent neurodegenerative diseases. Therapeutic strategies targeting lysosomes and autophagic machinery have already been tested in several of these diseases with promising results, suggesting that improving lysosomal function could be similarly beneficial in preserving function in the aging brain.
Collapse
Affiliation(s)
- Wesley Peng
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, 60611, USA
| | - Georgia Minakaki
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, 60611, USA
| | - Maria Nguyen
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, 60611, USA
| | - Dimitri Krainc
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, Illinois, 60611, USA.
| |
Collapse
|
44
|
Tellone E, Galtieri A, Ficarra S. Reviewing Biochemical Implications of Normal and Mutated Huntingtin in Huntington's Disease. Curr Med Chem 2019; 27:5137-5158. [PMID: 31223078 DOI: 10.2174/0929867326666190621101909] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2019] [Revised: 05/08/2019] [Accepted: 05/22/2019] [Indexed: 12/17/2022]
Abstract
Huntingtin (Htt) is a multi-function protein of the brain. Normal Htt shows a common alpha-helical structure but conformational changes in the form with beta strands are the principal cause of Huntington's disease. Huntington's disease is a genetic neurological disorder caused by a repeated expansion of the CAG trinucleotide, causing instability in the N-terminal of the gene coding for the Huntingtin protein. The mutation leads to the abnormal expansion of the production of the polyglutamine tract (polyQ) resulting in the form of an unstable Huntingtin protein commonly referred to as mutant Huntingtin. Mutant Huntingtin is the cause of the complex neurological metabolic alteration of Huntington's disease, resulting in both the loss of all the functions of normal Huntingtin and the genesis of abnormal interactions due to the presence of this mutation. One of the problems arising from the misfolded Huntingtin is the increase in oxidative stress, which is common in many neurological diseases such as Alzheimer's, Parkinson's, Amyotrophic Lateral Sclerosis and Creutzfeldt-Jakob disease. In the last few years, the use of antioxidants had a strong incentive to find valid therapies for defence against neurodegenerations. Although further studies are needed, the use of antioxidant mixtures to counteract neuronal damages seems promising.
Collapse
Affiliation(s)
- Ester Tellone
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, V.le Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Antonio Galtieri
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, V.le Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| | - Silvana Ficarra
- Department of Chemical, Biological, Pharmaceutical and Environmental Sciences, University of Messina, V.le Ferdinando Stagno d'Alcontres 31, 98166 Messina, Italy
| |
Collapse
|
45
|
Matlahov I, van der Wel PC. Conformational studies of pathogenic expanded polyglutamine protein deposits from Huntington's disease. Exp Biol Med (Maywood) 2019; 244:1584-1595. [PMID: 31203656 PMCID: PMC6920524 DOI: 10.1177/1535370219856620] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Huntington’s disease, like other neurodegenerative diseases, continues to lack an
effective cure. Current treatments that address early symptoms ultimately fail
Huntington’s disease patients and their families, with the disease typically
being fatal within 10–15 years from onset. Huntington’s disease is an inherited
disorder with motor and mental impairment, and is associated with the genetic
expansion of a CAG codon repeat encoding a polyglutamine-segment-containing
protein called huntingtin. These Huntington’s disease mutations cause misfolding
and aggregation of fragments of the mutant huntingtin protein, thereby likely
contributing to disease toxicity through a combination of gain-of-toxic-function
for the misfolded aggregates and a loss of function from sequestration of
huntingtin and other proteins. As with other amyloid diseases, the mutant
protein forms non-native fibrillar structures, which in Huntington’s disease are
found within patients’ neurons. The intracellular deposits are associated with
dysregulation of vital processes, and inter-neuronal transport of aggregates may
contribute to disease progression. However, a molecular understanding of these
aggregates and their detrimental effects has been frustrated by insufficient
structural data on the misfolded protein state. In this review, we examine
recent developments in the structural biology of polyglutamine-expanded
huntingtin fragments, and especially the contributions enabled by advances in
solid-state nuclear magnetic resonance spectroscopy. We summarize and discuss
our current structural understanding of the huntingtin deposits and how this
information furthers our understanding of the misfolding mechanism and disease
toxicity mechanisms.
Collapse
Affiliation(s)
- Irina Matlahov
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Patrick Ca van der Wel
- Department of Structural Biology, University of Pittsburgh, Pittsburgh, PA 15213, USA.,Zernike Institute for Advanced Materials, University of Groningen, Groningen 9747 AG, The Netherlands
| |
Collapse
|
46
|
Hartlage-Rübsamen M, Ratz V, Zeitschel U, Finzel L, Machner L, Köppen J, Schulze A, Demuth HU, von Hörsten S, Höfling C, Roßner S. Endogenous mouse huntingtin is highly abundant in cranial nerve nuclei, co-aggregates to Abeta plaques and is induced in reactive astrocytes in a transgenic mouse model of Alzheimer's disease. Acta Neuropathol Commun 2019; 7:79. [PMID: 31109380 PMCID: PMC6526682 DOI: 10.1186/s40478-019-0726-2] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2019] [Accepted: 04/17/2019] [Indexed: 12/11/2022] Open
Abstract
Pathogenic variants of the huntingtin (HTT) protein and their aggregation have been investigated in great detail in brains of Huntington's disease patients and HTT-transgenic animals. However, little is known about the physiological brain region- and cell type-specific HTT expression pattern in wild type mice and a potential recruitment of endogenous HTT to other pathogenic protein aggregates such as amyloid plaques in cross seeding events. Employing a monoclonal anti-HTT antibody directed against the HTT mid-region and using brain tissue of three different mouse strains, we detected prominent immunoreactivity in a number of brain areas, particularly in cholinergic cranial nerve nuclei, while ubiquitous neuronal staining appeared faint. The region-specific distribution of endogenous HTT was found to be comparable in wild type rat and hamster brain. In human amyloid precursor protein transgenic Tg2576 mice with amyloid plaque pathology, similar neuronal HTT expression patterns and a distinct association of HTT with Abeta plaques were revealed by immunohistochemical double labelling. Additionally, the localization of HTT in reactive astrocytes was demonstrated for the first time in a transgenic Alzheimer's disease animal model. Both, plaque association of HTT and occurrence in astrocytes appeared to be age-dependent. Astrocytic HTT gene and protein expression was confirmed in primary cultures by RT-qPCR and by immunocytochemistry. We provide the first detailed analysis of physiological HTT expression in rodent brain and, under pathological conditions, demonstrate HTT aggregation in proximity to Abeta plaques and Abeta-induced astrocytic expression of endogenous HTT in Tg2576 mice.
Collapse
Affiliation(s)
| | - Veronika Ratz
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Preclinical Experimental Center, Erlangen, Germany
| | - Ulrike Zeitschel
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Lukas Finzel
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Lisa Machner
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Molecular Drug Design and Target Validation, Halle (Saale), Germany
| | - Janett Köppen
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Molecular Drug Design and Target Validation, Halle (Saale), Germany
| | - Anja Schulze
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Molecular Drug Design and Target Validation, Halle (Saale), Germany
| | - Hans-Ulrich Demuth
- Fraunhofer Institute for Cell Therapy and Immunology, Department of Molecular Drug Design and Target Validation, Halle (Saale), Germany
| | - Stephan von Hörsten
- Friedrich-Alexander-Universität Erlangen-Nürnberg, Preclinical Experimental Center, Erlangen, Germany
| | - Corinna Höfling
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany
| | - Steffen Roßner
- Paul Flechsig Institute for Brain Research, University of Leipzig, Leipzig, Germany.
| |
Collapse
|
47
|
Olfactory bulb atrophy and caspase activation observed in the BACHD rat models of Huntington disease. Neurobiol Dis 2019; 125:219-231. [DOI: 10.1016/j.nbd.2019.02.002] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2018] [Revised: 12/14/2018] [Accepted: 02/04/2019] [Indexed: 01/08/2023] Open
|
48
|
Couly S, Paucard A, Bonneaud N, Maurice T, Benigno L, Jourdan C, Cohen-Solal C, Vignes M, Maschat F. Improvement of BDNF signalling by P42 peptide in Huntington's disease. Hum Mol Genet 2019; 27:3012-3028. [PMID: 29860423 DOI: 10.1093/hmg/ddy207] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2018] [Accepted: 05/23/2018] [Indexed: 12/21/2022] Open
Abstract
Huntington's disease (HD) is caused by a mutation in the Huntingtin (HTT) protein. We previously reported that the 23aa peptide of HTT protein, P42, is preventing HD pathological phenotypes, such as aggregation, reduction of motor performances and neurodegeneration. A systemic treatment with P42 during the pre-symptomatic phase of the disease showed therapeutic potential in R6/2 mice. We here tested P42 effects when administered during the post-symptomatic phase. The P42 treatment alleviated deficits in motor performances, even when symptoms have already started. Because changes in the level and activity of brain-derived neurotrophic factor (BDNF) have been shown to play a central role in HD, we analysed the influence of P42 on BDNF deficit and associated phenotypes. Our data suggest that P42 is involved in the spatio-temporal control of bdnf and trkB mRNA and their protein levels. Related to this enhancement of BDNF-TrkB signalling, R6/2 mice treated with P42, exhibit reduced anxiety, better learning and memory performances, and better long-term potentiation (LTP) response. Finally we identified a direct influence of P42 peptide on neuronal plasticity and activity. These results suggest that P42 offers an efficient therapeutic potential not only by preventing aggregation of mutant HTT at early stages of the disease, but also by favouring some physiological functions of normal HTT, as P42 is naturally part of it, at the different stages of the disease. This makes P42 peptide potentially suitable not only to prevent, but also to treat HD.
Collapse
Affiliation(s)
- Simon Couly
- MMDN, Univ-Montpellier, EPHE, INSERM, UMR-S1198, Montpellier F-34095, France
| | - Alexia Paucard
- MMDN, Univ-Montpellier, EPHE, INSERM, UMR-S1198, Montpellier F-34095, France
| | - Nathalie Bonneaud
- MMDN, Univ-Montpellier, EPHE, INSERM, UMR-S1198, Montpellier F-34095, France
| | - Tangui Maurice
- MMDN, Univ-Montpellier, EPHE, INSERM, UMR-S1198, Montpellier F-34095, France
| | | | - Christophe Jourdan
- MMDN, Univ-Montpellier, EPHE, INSERM, UMR-S1198, Montpellier F-34095, France
| | | | - Michel Vignes
- IBMM-UMR5247, Univ-Montpellier, Montpellier F-34095, France
| | - Florence Maschat
- MMDN, Univ-Montpellier, EPHE, INSERM, UMR-S1198, Montpellier F-34095, France
| |
Collapse
|
49
|
Weber JJ, Kloock SJ, Nagel M, Ortiz-Rios MM, Hofmann J, Riess O, Nguyen HP. Calpastatin ablation aggravates the molecular phenotype in cell and animal models of Huntington disease. Neuropharmacology 2018; 133:94-106. [PMID: 29355642 DOI: 10.1016/j.neuropharm.2018.01.022] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2017] [Revised: 12/21/2017] [Accepted: 01/16/2018] [Indexed: 12/15/2022]
Abstract
Deciphering the molecular pathology of Huntington disease is of particular importance, not only for a better understanding of this neurodegenerative disease, but also to identify potential therapeutic targets. The polyglutamine-expanded disease protein huntingtin was shown to undergo proteolysis, which results in the accumulation of toxic and aggregation-prone fragments. Amongst several classes of proteolytic enzymes responsible for huntingtin processing, the group of calcium-activated calpains has been found to be a significant mediator of the disease protein toxicity. To confirm the impact of calpain-mediated huntingtin cleavage in Huntington disease, we analysed the effect of depleting or overexpressing the endogenous calpain inhibitor calpastatin in HEK293T cells transfected with wild-type or polyglutamine-expanded huntingtin. Moreover, we crossbred huntingtin knock-in mice with calpastatin knockout animals to assess its effect not only on huntingtin cleavage and aggregation but also additional molecular markers. We demonstrated that a reduced or ablated expression of calpastatin triggers calpain overactivation and a consequently increased mutant huntingtin cleavage in cells and in vivo. These alterations were accompanied by an elevated formation of predominantly cytoplasmic huntingtin aggregates. On the other hand, overexpression of calpastatin in cells attenuated huntingtin fragmentation and aggregation. In addition, we observed an enhanced cleavage of DARPP-32, p35 and synapsin-1 in neuronal tissue upon calpain overactivation. Our results corroborate the important role of calpains in the molecular pathogenesis of Huntington disease and endorse targeting these proteolytic enzymes as a therapeutic approach.
Collapse
Affiliation(s)
- Jonasz Jeremiasz Weber
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstraße 7, 72076, Tübingen, Germany; Centre for Rare Diseases, University of Tübingen, Calwerstraße 7, 72076, Tübingen, Germany.
| | - Simon Johannes Kloock
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstraße 7, 72076, Tübingen, Germany; Centre for Rare Diseases, University of Tübingen, Calwerstraße 7, 72076, Tübingen, Germany.
| | - Maike Nagel
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstraße 7, 72076, Tübingen, Germany; Centre for Rare Diseases, University of Tübingen, Calwerstraße 7, 72076, Tübingen, Germany.
| | - Midea Malena Ortiz-Rios
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstraße 7, 72076, Tübingen, Germany; Centre for Rare Diseases, University of Tübingen, Calwerstraße 7, 72076, Tübingen, Germany.
| | - Julian Hofmann
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstraße 7, 72076, Tübingen, Germany; Centre for Rare Diseases, University of Tübingen, Calwerstraße 7, 72076, Tübingen, Germany.
| | - Olaf Riess
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstraße 7, 72076, Tübingen, Germany; Centre for Rare Diseases, University of Tübingen, Calwerstraße 7, 72076, Tübingen, Germany.
| | - Huu Phuc Nguyen
- Institute of Medical Genetics and Applied Genomics, University of Tübingen, Calwerstraße 7, 72076, Tübingen, Germany; Centre for Rare Diseases, University of Tübingen, Calwerstraße 7, 72076, Tübingen, Germany.
| |
Collapse
|
50
|
Croce KR, Yamamoto A. A role for autophagy in Huntington's disease. Neurobiol Dis 2018; 122:16-22. [PMID: 30149183 DOI: 10.1016/j.nbd.2018.08.010] [Citation(s) in RCA: 96] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2018] [Revised: 08/10/2018] [Accepted: 08/23/2018] [Indexed: 12/19/2022] Open
Abstract
The lysosome-mediated degradation pathway known as macroautophagy is the most versatile means through which cells can eliminate and recycle unwanted materials. Through both selective and non-selective means, macroautophagy can degrade a wide range of cargoes from bulk cytosol to organelles and aggregated proteins. Although studies of disorders such as Parkinson's disease and Amyotrophic Lateral Sclerosis suggest that autophagic and lysosomal dysfunction directly contributes to disease, this had not been the case for the polyglutamine disorder Huntington's disease (HD), for which there was little indication of a disruption in the autophagic-lysosomal system. This supported the possibility of targeting autophagy as a much needed therapeutic approach to combat this disease. Possibly challenging this view, however, are a recent set of studies suggesting that the protein affected in Huntington's disease, huntingtin, might mechanistically contribute to macroautophagy. In this review, we will explore how autophagy might impact or be impacted by HD pathogenesis, and whether a therapeutic approach centering on autophagy may be possible for this yet incurable disease.
Collapse
Affiliation(s)
- Katherine R Croce
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, United States
| | - Ai Yamamoto
- Department of Pathology and Cell Biology, Columbia University, New York, NY 10032, United States; Department of Neurology, Columbia University, New York, NY 10032, United States.
| |
Collapse
|