1
|
Harding RJ, Xie Y, Caron NS, Findlay-Black H, Lyu C, Potluri N, Chandrasekaran R, Hayden MR, Leavitt BR, Langbehn DR, Southwell AL. Challenges and advances for huntingtin detection in cerebrospinal fluid: in support of relative quantification. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2024:2024.09.25.614766. [PMID: 39386513 PMCID: PMC11463412 DOI: 10.1101/2024.09.25.614766] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/12/2024]
Abstract
Huntington disease (HD) is a progressive and devastating neurodegenerative disease caused by expansion of a glutamine-coding CAG tract in the huntingtin (HTT) gene above a critical threshold of ~35 repeats resulting in expression of mutant HTT (mHTT). A promising treatment approach being tested in clinical trials is HTT lowering, which aims to reduce levels of the mHTT protein. Target engagement of these therapies in the brain are inferred using antibody-based assays to measure mHTT levels in the cerebrospinal fluid (CSF), which is frequently reported as absolute mHTT concentration based on a monomeric protein standard used to generate a standard curve. However, patient biofluids are a complex milieu of different mHTT protein species, suggesting that absolute quantitation is challenging, and a single, recombinant protein standard may not be sufficient to interpret assay signal as molar mHTT concentration. In this study, we used immunoprecipitation and flow cytometry (IP-FCM) to investigate different factors that influence mHTT detection assay signal. Our results show that HTT protein fragmentation, protein-protein interactions, affinity tag positioning, oligomerization and polyglutamine tract length affect assay signal intensity, indicating that absolute HTT quantitation in heterogeneous biological samples is not possible with current technologies using a single standard protein. We also explore the binding specificity of the MW1 anti-polyglutamine antibody, commonly used in these assays as a mHTT-selective reagent and demonstrate that mHTT binding is preferred but not specific. Furthermore, we find that MW1 depletion is not only incomplete, leaving residual mHTT, but also non-specific, resulting in pull down of some wildtype HTT protein. Based on these observations, we recommend that mHTT detection assays report only relative mHTT quantitation using normalized arbitrary units of assay signal intensity, rather than molar concentrations, in the assessment of central nervous system HTT lowering in ongoing clinical and preclinical studies, and that MW1-depletion not be used a method for quantifying wildtype HTT protein.
Collapse
Affiliation(s)
- Rachel J. Harding
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
- Structural Genomics Consortium, University of Toronto, Toronto, ON M5G 1L7, Canada
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Yuanyun Xie
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32827, USA
| | - Nicholas S. Caron
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC V5Z 4H4 Canada
| | - Hailey Findlay-Black
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC V5Z 4H4 Canada
| | - Caroline Lyu
- Structural Genomics Consortium, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Nalini Potluri
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32827, USA
| | - Renu Chandrasekaran
- Leslie Dan Faculty of Pharmacy, University of Toronto, Toronto, ON M5S 3M2, Canada
| | - Michael R. Hayden
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC V5Z 4H4 Canada
| | - Blair R. Leavitt
- Centre for Molecular Medicine and Therapeutics, The University of British Columbia, Vancouver, BC V5Z 4H4 Canada
| | - Douglas R. Langbehn
- Department of Psychiatry, Carver College of Medicine, University of Iowa, Iowa City, IA 52242, USA
| | - Amber L. Southwell
- Burnett School of Biomedical Sciences, University of Central Florida, Orlando, FL, 32827, USA
| |
Collapse
|
2
|
Ramazi S, Dadzadi M, Darvazi M, Seddigh N, Allahverdi A. Protein modification in neurodegenerative diseases. MedComm (Beijing) 2024; 5:e674. [PMID: 39105197 PMCID: PMC11298556 DOI: 10.1002/mco2.674] [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: 12/05/2023] [Revised: 07/02/2024] [Accepted: 07/08/2024] [Indexed: 08/07/2024] Open
Abstract
Posttranslational modifications play a crucial role in governing cellular functions and protein behavior. Researchers have implicated dysregulated posttranslational modifications in protein misfolding, which results in cytotoxicity, particularly in neurodegenerative diseases such as Alzheimer disease, Parkinson disease, and Huntington disease. These aberrant posttranslational modifications cause proteins to gather in certain parts of the brain that are linked to the development of the diseases. This leads to neuronal dysfunction and the start of neurodegenerative disease symptoms. Cognitive decline and neurological impairments commonly manifest in neurodegenerative disease patients, underscoring the urgency of comprehending the posttranslational modifications' impact on protein function for targeted therapeutic interventions. This review elucidates the critical link between neurodegenerative diseases and specific posttranslational modifications, focusing on Tau, APP, α-synuclein, Huntingtin protein, Parkin, DJ-1, and Drp1. By delineating the prominent aberrant posttranslational modifications within Alzheimer disease, Parkinson disease, and Huntington disease, the review underscores the significance of understanding the interplay among these modifications. Emphasizing 10 key abnormal posttranslational modifications, this study aims to provide a comprehensive framework for investigating neurodegenerative diseases holistically. The insights presented herein shed light on potential therapeutic avenues aimed at modulating posttranslational modifications to mitigate protein aggregation and retard neurodegenerative disease progression.
Collapse
Affiliation(s)
- Shahin Ramazi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Maedeh Dadzadi
- Department of BiotechnologyFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Mona Darvazi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| | - Nasrin Seddigh
- Department of BiochemistryFaculty of Advanced Science and TechnologyTehran Medical SciencesIslamic Azad UniversityTehranIran
| | - Abdollah Allahverdi
- Department of BiophysicsFaculty of Biological SciencesTarbiat Modares UniversityTehranIran
| |
Collapse
|
3
|
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
|
4
|
Azam HMH, Rößling RI, Geithe C, Khan MM, Dinter F, Hanack K, Prüß H, Husse B, Roggenbuck D, Schierack P, Rödiger S. MicroRNA biomarkers as next-generation diagnostic tools for neurodegenerative diseases: a comprehensive review. Front Mol Neurosci 2024; 17:1386735. [PMID: 38883980 PMCID: PMC11177777 DOI: 10.3389/fnmol.2024.1386735] [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: 02/15/2024] [Accepted: 04/12/2024] [Indexed: 06/18/2024] Open
Abstract
Neurodegenerative diseases (NDs) are characterized by abnormalities within neurons of the brain or spinal cord that gradually lose function, eventually leading to cell death. Upon examination of affected tissue, pathological changes reveal a loss of synapses, misfolded proteins, and activation of immune cells-all indicative of disease progression-before severe clinical symptoms become apparent. Early detection of NDs is crucial for potentially administering targeted medications that may delay disease advancement. Given their complex pathophysiological features and diverse clinical symptoms, there is a pressing need for sensitive and effective diagnostic methods for NDs. Biomarkers such as microRNAs (miRNAs) have been identified as potential tools for detecting these diseases. We explore the pivotal role of miRNAs in the context of NDs, focusing on Alzheimer's disease, Parkinson's disease, Multiple sclerosis, Huntington's disease, and Amyotrophic Lateral Sclerosis. The review delves into the intricate relationship between aging and NDs, highlighting structural and functional alterations in the aging brain and their implications for disease development. It elucidates how miRNAs and RNA-binding proteins are implicated in the pathogenesis of NDs and underscores the importance of investigating their expression and function in aging. Significantly, miRNAs exert substantial influence on post-translational modifications (PTMs), impacting not just the nervous system but a wide array of tissues and cell types as well. Specific miRNAs have been found to target proteins involved in ubiquitination or de-ubiquitination processes, which play a significant role in regulating protein function and stability. We discuss the link between miRNA, PTM, and NDs. Additionally, the review discusses the significance of miRNAs as biomarkers for early disease detection, offering insights into diagnostic strategies.
Collapse
Affiliation(s)
- Hafiz Muhammad Husnain Azam
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Rosa Ilse Rößling
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Christiane Geithe
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, The Brandenburg Medical School Theodor Fontane and the University of Potsdam, Berlin, Germany
| | - Muhammad Moman Khan
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Franziska Dinter
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
- PolyAn GmbH, Berlin, Germany
| | - Katja Hanack
- Institute of Biochemistry and Biology, University of Potsdam, Potsdam, Germany
| | - Harald Prüß
- German Center for Neurodegenerative Diseases (DZNE), Berlin, Germany
- Department of Neurology, Charité - Universitätsmedizin Berlin, Berlin, Germany
| | - Britta Husse
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Dirk Roggenbuck
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Peter Schierack
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
| | - Stefan Rödiger
- Institute of Biotechnology, Faculty of Environment and Natural Sciences, Brandenburg University of Technology Cottbus-Senftenberg, Senftenberg, Germany
- Faculty of Health Sciences, Joint Faculty of the Brandenburg University of Technology Cottbus - Senftenberg, The Brandenburg Medical School Theodor Fontane and the University of Potsdam, Berlin, Germany
| |
Collapse
|
5
|
Thomson SB, Stam A, Brouwers C, Fodale V, Bresciani A, Vermeulen M, Mostafavi S, Petkau TL, Hill A, Yung A, Russell-Schulz B, Kozlowski P, MacKay A, Ma D, Beg MF, Evers MM, Vallès A, Leavitt BR. AAV5-miHTT-mediated huntingtin lowering improves brain health in a Huntington's disease mouse model. Brain 2023; 146:2298-2315. [PMID: 36508327 PMCID: PMC10232253 DOI: 10.1093/brain/awac458] [Citation(s) in RCA: 6] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 06/23/2022] [Accepted: 10/30/2022] [Indexed: 04/12/2024] Open
Abstract
Huntingtin (HTT)-lowering therapies show great promise in treating Huntington's disease. We have developed a microRNA targeting human HTT that is delivered in an adeno-associated serotype 5 viral vector (AAV5-miHTT), and here use animal behaviour, MRI, non-invasive proton magnetic resonance spectroscopy and striatal RNA sequencing as outcome measures in preclinical mouse studies of AAV5-miHTT. The effects of AAV5-miHTT treatment were evaluated in homozygous Q175FDN mice, a mouse model of Huntington's disease with severe neuropathological and behavioural phenotypes. Homozygous mice were used instead of the more commonly used heterozygous strain, which exhibit milder phenotypes. Three-month-old homozygous Q175FDN mice, which had developed acute phenotypes by the time of treatment, were injected bilaterally into the striatum with either formulation buffer (phosphate-buffered saline + 5% sucrose), low dose (5.2 × 109 genome copies/mouse) or high dose (1.3 × 1011 genome copies/mouse) AAV5-miHTT. Wild-type mice injected with formulation buffer served as controls. Behavioural assessments of cognition, T1-weighted structural MRI and striatal proton magnetic resonance spectroscopy were performed 3 months after injection, and shortly afterwards the animals were sacrificed to collect brain tissue for protein and RNA analysis. Motor coordination was assessed at 1-month intervals beginning at 2 months of age until sacrifice. Dose-dependent changes in AAV5 vector DNA level, miHTT expression and mutant HTT were observed in striatum and cortex of AAV5-miHTT-treated Huntington's disease model mice. This pattern of microRNA expression and mutant HTT lowering rescued weight loss in homozygous Q175FDN mice but did not affect motor or cognitive phenotypes. MRI volumetric analysis detected atrophy in four brain regions in homozygous Q175FDN mice, and treatment with high dose AAV5-miHTT rescued this effect in the hippocampus. Like previous magnetic resonance spectroscopy studies in Huntington's disease patients, decreased total N-acetyl aspartate and increased myo-inositol levels were found in the striatum of homozygous Q175FDN mice. These neurochemical findings were partially reversed with AAV5-miHTT treatment. Striatal transcriptional analysis using RNA sequencing revealed mutant HTT-induced changes that were partially reversed by HTT lowering with AAV5-miHTT. Striatal proton magnetic resonance spectroscopy analysis suggests a restoration of neuronal function, and striatal RNA sequencing analysis shows a reversal of transcriptional dysregulation following AAV5-miHTT in a homozygous Huntington's disease mouse model with severe pathology. The results of this study support the use of magnetic resonance spectroscopy in HTT-lowering clinical trials and strengthen the therapeutic potential of AAV5-miHTT in reversing severe striatal dysfunction in Huntington's disease.
Collapse
Affiliation(s)
- Sarah B Thomson
- Department of Medical Genetics, Centre for Molecular Medicine & Therapeutics, University of British Columbia and BC Children’s Hospital, Vancouver, BC V5Z4H4, Canada
| | - Anouk Stam
- Department of Research & Development, uniQure Biopharma B.V., 1105BP Amsterdam, The Netherlands
| | - Cynthia Brouwers
- Department of Research & Development, uniQure Biopharma B.V., 1105BP Amsterdam, The Netherlands
| | - Valentina Fodale
- Department of Translational Biology, IRBM S.p.A., Pomezia 00071, Italy
| | - Alberto Bresciani
- Department of Translational Biology, IRBM S.p.A., Pomezia 00071, Italy
| | - Michael Vermeulen
- Department of Medical Genetics, Centre for Molecular Medicine & Therapeutics, University of British Columbia and BC Children’s Hospital, Vancouver, BC V5Z4H4, Canada
| | - Sara Mostafavi
- Department of Medical Genetics, Centre for Molecular Medicine & Therapeutics, University of British Columbia and BC Children’s Hospital, Vancouver, BC V5Z4H4, Canada
| | - Terri L Petkau
- Department of Medical Genetics, Centre for Molecular Medicine & Therapeutics, University of British Columbia and BC Children’s Hospital, Vancouver, BC V5Z4H4, Canada
| | - Austin Hill
- Department of Medical Genetics, Centre for Molecular Medicine & Therapeutics, University of British Columbia and BC Children’s Hospital, Vancouver, BC V5Z4H4, Canada
| | - Andrew Yung
- UBC MRI Research Centre, Department of Radiology, University of British Columbia, Vancouver, BC V6T2B5, Canada
| | - Bretta Russell-Schulz
- UBC MRI Research Centre, Department of Radiology, University of British Columbia, Vancouver, BC V6T2B5, Canada
| | - Piotr Kozlowski
- UBC MRI Research Centre, Department of Radiology, University of British Columbia, Vancouver, BC V6T2B5, Canada
| | - Alex MacKay
- UBC MRI Research Centre, Department of Radiology, University of British Columbia, Vancouver, BC V6T2B5, Canada
| | - Da Ma
- Wake Forest University School of Medicine, Medical Center Boulevard, Winston-Salem, NC 27101, USA
| | - Mirza Faisal Beg
- School of Engineering Science, Simon Fraser University, Burnaby, BC V5A0A7, Canada
| | - Melvin M Evers
- Department of Research & Development, uniQure Biopharma B.V., 1105BP Amsterdam, The Netherlands
| | - Astrid Vallès
- Department of Research & Development, uniQure Biopharma B.V., 1105BP Amsterdam, The Netherlands
| | - Blair R Leavitt
- Department of Medical Genetics, Centre for Molecular Medicine & Therapeutics, University of British Columbia and BC Children’s Hospital, Vancouver, BC V5Z4H4, Canada
| |
Collapse
|
6
|
Cyclodextrin-Based Nanoparticles for Delivery of Antisense Oligonucleotides Targeting Huntingtin. Pharmaceutics 2023; 15:pharmaceutics15020520. [PMID: 36839842 PMCID: PMC9965918 DOI: 10.3390/pharmaceutics15020520] [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: 12/21/2022] [Revised: 01/10/2023] [Accepted: 02/02/2023] [Indexed: 02/09/2023] Open
Abstract
Huntington's disease (HD) is a progressive inherited neurodegenerative disease caused by a CAG repeat expansion in the huntingtin gene, which is translated into the pathologic mutant huntingtin (mHTT) protein. Despite the great potential of HTT lowering strategies and the numerous antisense oligonucleotides (ASOs) in pre- and clinical trials, sustained silencing of mHTT has not been achieved. As a strategy to improve ASO delivery, cyclodextrin-based nanoparticles (CDs) offer a promising approach. Here, three CDs with distinct chemical structures were designed and their efficacies were compared as potential platforms for the delivery of ASO targeting HTT. Results using striatal neurons and HD patient-derived fibroblasts indicate that modified γ-CDs exhibited the best uptake efficiency and successfully downregulated mHTT at protein and allele levels. The incorporation of the brain-targeting peptide RVG into the modified γ-CDs showed greater downregulation of mHTT protein and HD-causing allele SNP1 than untargeted ones in an in vitro blood-brain barrier model. Although the ASO sequence was designed as a nonallele-specific therapeutic approach, our strategy gives an additional benefit of some mHTT selectivity. Overall, this study demonstrated the CD platform's feasibility for delivering ASO-based therapeutics for HD treatment.
Collapse
|
7
|
Joachimiak P, Ciesiołka A, Kozłowska E, Świtoński PM, Figura G, Ciołak A, Adamek G, Surdyka M, Kalinowska-Pośka Ż, Figiel M, Caron NS, Hayden MR, Fiszer A. Allele-specific quantitation of ATXN3 and HTT transcripts in polyQ disease models. BMC Biol 2023; 21:17. [PMID: 36726088 PMCID: PMC9893648 DOI: 10.1186/s12915-023-01515-3] [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: 09/06/2022] [Accepted: 01/17/2023] [Indexed: 02/03/2023] Open
Abstract
BACKGROUND The majority of genes in the human genome is present in two copies but the expression levels of both alleles is not equal. Allelic imbalance is an aspect of gene expression relevant not only in the context of genetic variation, but also to understand the pathophysiology of genes implicated in genetic disorders, in particular, dominant genetic diseases where patients possess one normal and one mutant allele. Polyglutamine (polyQ) diseases are caused by the expansion of CAG trinucleotide tracts within specific genes. Spinocerebellar ataxia type 3 (SCA3) and Huntington's disease (HD) patients harbor one normal and one mutant allele that differ in the length of CAG tracts. However, assessing the expression level of individual alleles is challenging due to the presence of abundant CAG repeats in the human transcriptome, which make difficult the design of allele-specific methods, as well as of therapeutic strategies to selectively engage CAG sequences in mutant transcripts. RESULTS To precisely quantify expression in an allele-specific manner, we used SNP variants that are linked to either normal or CAG expanded alleles of the ataxin-3 (ATXN3) and huntingtin (HTT) genes in selected patient-derived cell lines. We applied a SNP-based quantitative droplet digital PCR (ddPCR) protocol for precise determination of the levels of transcripts in cellular and mouse models. For HD, we showed that the process of cell differentiation can affect the ratio between endogenous alleles of HTT mRNA. Additionally, we reported changes in the absolute number of the ATXN3 and HTT transcripts per cell during neuronal differentiation. We also implemented our assay to reliably monitor, in an allele-specific manner, the silencing efficiency of mRNA-targeting therapeutic approaches for HD. Finally, using the humanized Hu128/21 HD mouse model, we showed that the ratio of normal and mutant HTT transgene expression in brain slightly changes with the age of mice. CONCLUSIONS Using allele-specific ddPCR assays, we observed differences in allele expression levels in the context of SCA3 and HD. Our allele-selective approach is a reliable and quantitative method to analyze low abundant transcripts and is performed with high accuracy and reproducibility. Therefore, the use of this approach can significantly improve understanding of allele-related mechanisms, e.g., related with mRNA processing that may be affected in polyQ diseases.
Collapse
Affiliation(s)
- Paweł Joachimiak
- grid.413454.30000 0001 1958 0162Department of Medical Biotechnology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Adam Ciesiołka
- grid.413454.30000 0001 1958 0162Department of Medical Biotechnology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Emilia Kozłowska
- grid.413454.30000 0001 1958 0162Department of Medical Biotechnology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Paweł M. Świtoński
- grid.413454.30000 0001 1958 0162Department of Medical Biotechnology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Grzegorz Figura
- grid.413454.30000 0001 1958 0162Department of Medical Biotechnology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Agata Ciołak
- grid.413454.30000 0001 1958 0162Department of Medical Biotechnology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Grażyna Adamek
- grid.413454.30000 0001 1958 0162Department of Medical Biotechnology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Magdalena Surdyka
- grid.413454.30000 0001 1958 0162Department of Molecular Neurobiology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Żaneta Kalinowska-Pośka
- grid.413454.30000 0001 1958 0162Department of Molecular Neurobiology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Maciej Figiel
- grid.413454.30000 0001 1958 0162Department of Molecular Neurobiology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| | - Nicholas S. Caron
- grid.17091.3e0000 0001 2288 9830Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC V5Z 4H4 Canada
| | - Michael R. Hayden
- grid.17091.3e0000 0001 2288 9830Centre for Molecular Medicine and Therapeutics, BC Children’s Hospital Research Institute, Department of Medical Genetics, University of British Columbia, Vancouver, BC V5Z 4H4 Canada
| | - Agnieszka Fiszer
- grid.413454.30000 0001 1958 0162Department of Medical Biotechnology, Institute of Bioorganic Chemistry, Polish Academy of Sciences, Noskowskiego 12/14, 61-704 Poznan, Poland
| |
Collapse
|
8
|
Parkin GM, Corey-Bloom J, Snell C, Smith H, Laurenza A, Daldin M, Bresciani A, Thomas EA. Salivary Huntingtin protein is uniquely associated with clinical features of Huntington's disease. Sci Rep 2023; 13:1034. [PMID: 36658243 PMCID: PMC9852574 DOI: 10.1038/s41598-023-28019-y] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Accepted: 01/11/2023] [Indexed: 01/20/2023] Open
Abstract
Measuring Huntingtin (HTT) protein in peripheral cells represents an essential step in biomarker discovery for Huntington's Disease (HD), however to date, investigations into the salivary expression of HTT has been lacking. In the current study, we quantified total HTT (tHTT) and mutant HTT (mHTT) protein in matched blood and saliva samples using single molecule counting (SMC) immunoassays: 2B7-D7F7 (tHTT) and 2B7-MW1 (mHTT). Matched samples, and clinical data, were collected from 95 subjects: n = 19 manifest HD, n = 34 premanifest HD (PM), and n = 42 normal controls (NC). Total HTT and mHTT levels were not correlated in blood and saliva. Plasma tHTT was significantly associated with age, and participant sex; whereas salivary mHTT was significantly correlated with age, CAG repeat length and CAP score. Plasma and salivary tHTT did not differ across cohorts. Salivary and plasma mHTT were significantly increased in PM compared to NC; salivary mHTT was also significantly increased in HD compared to NC. Only salivary tHTT and mHTT were significantly correlated with clinical measures. Salivary HTT is uniquely associated with clinical measures of HD and offers significant promise as a relevant, non-invasive HD biomarker. Its use could be immediately implemented into both translational and clinical research applications.
Collapse
Affiliation(s)
- Georgia M Parkin
- Department of Epidemiology, University of California Irvine, Irvine, CA, USA.
- Institute for Interdisciplinary Salivary Bioscience Research, University of California Irvine, Irvine, CA, USA.
| | - Jody Corey-Bloom
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA
| | - Chase Snell
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA
| | - Haileigh Smith
- Department of Neurosciences, University of California San Diego, San Diego, CA, USA
| | - Angela Laurenza
- Department of Translational Biology, IRBM S.p.A., via Pontina Km 30, 600, Pomezia, Rome, Italy
- Menarini Ricerche S.p.A., via Tito Speri 10, Pomezia, Rome, Italy
| | - Manuel Daldin
- Department of Translational Biology, IRBM S.p.A., via Pontina Km 30, 600, Pomezia, Rome, Italy
| | - Alberto Bresciani
- Department of Translational Biology, IRBM S.p.A., via Pontina Km 30, 600, Pomezia, Rome, Italy
- Exscientia, Oxford Science Park, Oxford, UK
| | - Elizabeth A Thomas
- Department of Epidemiology, University of California Irvine, Irvine, CA, USA
- Institute for Interdisciplinary Salivary Bioscience Research, University of California Irvine, Irvine, CA, USA
| |
Collapse
|
9
|
Pepe G, Capocci L, Marracino F, Realini N, Lenzi P, Martinello K, Bovier TF, Bichell TJ, Scarselli P, Di Cicco C, Bowman AB, Digilio FA, Fucile S, Fornai F, Armirotti A, Parlato R, Di Pardo A, Maglione V. Treatment with THI, an inhibitor of sphingosine-1-phosphate lyase, modulates glycosphingolipid metabolism and results therapeutically effective in experimental models of Huntington's disease. Mol Ther 2023; 31:282-299. [PMID: 36116006 PMCID: PMC9840122 DOI: 10.1016/j.ymthe.2022.09.004] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2022] [Revised: 08/05/2022] [Accepted: 09/06/2022] [Indexed: 02/06/2023] Open
Abstract
Huntington's disease (HD) is a fatal neurodegenerative disorder with no effective cure currently available. Over the past few years our research has shown that alterations in sphingolipid metabolism represent a critical determinant in HD pathogenesis. In particular, aberrant metabolism of sphingosine-1-phosphate (S1P) has been reported in multiple disease settings, including human postmortem brains from HD patients. In this study, we investigate the potential therapeutic effect of the inhibition of S1P degradative enzyme SGPL1, by the chronic administration of the 2-acetyl-5-tetrahydroxybutyl imidazole (THI) inhibitor. We show that THI mitigated motor dysfunctions in both mouse and fly models of HD. The compound evoked the activation of pro-survival pathways, normalized levels of brain-derived neurotrophic factor, preserved white matter integrity, and stimulated synaptic functions in HD mice. Metabolically, THI restored normal levels of hexosylceramides and stimulated the autophagic and lysosomal machinery, facilitating the reduction of nuclear inclusions of both wild-type and mutant huntingtin proteins.
Collapse
Affiliation(s)
| | | | | | - Natalia Realini
- Analytical Chemistry Lab, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Paola Lenzi
- Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | | | - Tiziana Francesca Bovier
- Research Institute on Terrestrial Ecosystems (IRET), UOS Naples-CNR, Via Pietro Castellino 111, 80131 Naples, Italy; Department of Pediatrics Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA; Center for Host-Pathogen Interaction, Columbia University Vagelos College of Physicians and Surgeons, New York 10032, NY, USA
| | - Terry Jo Bichell
- Vanderbilt Brain Institute, Vanderbilt University, Nashville, TN 37240, USA
| | | | | | - Aaron B Bowman
- School of Health Sciences, Purdue University, West Lafayette, IN 47907-2051, USA
| | - Filomena A Digilio
- Research Institute on Terrestrial Ecosystems (IRET), UOS Naples-CNR, Via Pietro Castellino 111, 80131 Naples, Italy
| | - Sergio Fucile
- IRCCS Neuromed, Pozzilli (IS) 86077, Italy; Department of Physiology and Pharmacology, Sapienza Rome University, Rome 00185, Italy
| | - Francesco Fornai
- IRCCS Neuromed, Pozzilli (IS) 86077, Italy; Department of Translational Research and New Technologies in Medicine and Surgery, University of Pisa, Via Roma 55, 56126 Pisa, Italy
| | - Andrea Armirotti
- Analytical Chemistry Lab, Fondazione Istituto Italiano di Tecnologia, Via Morego 30, 16163 Genoa, Italy
| | - Rosanna Parlato
- Division for Neurodegenerative Diseases, Department of Neurology, Mannheim Center for Translational Neuroscience, Medical Faculty Mannheim Heidelberg University, Mannheim 68167, Germany
| | | | | |
Collapse
|
10
|
Galyan SM, Ewald CY, Jalencas X, Masrani S, Meral S, Mestres J. Fragment-based virtual screening identifies a first-in-class preclinical drug candidate for Huntington's disease. Sci Rep 2022; 12:19642. [PMID: 36385140 PMCID: PMC9668931 DOI: 10.1038/s41598-022-21900-2] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2022] [Accepted: 10/05/2022] [Indexed: 11/17/2022] Open
Abstract
Currently, there are no therapies available to modify the disease progression of Huntington's disease (HD). Recent clinical trial failures of antisense oligonucleotide candidates in HD have demonstrated the need for new therapeutic approaches. Here, we developed a novel in-silico fragment scanning approach across the surface of mutant huntingtin (mHTT) polyQ and predicted four hit compounds. Two rounds of compound analoging using a strategy of testing structurally similar compounds in an affinity assay rapidly identified GLYN122. In vitro, GLYN122 directly binds and reduces mHTT and induces autophagy in neurons. In vivo, our results confirm that GLYN122 can reduce mHTT in the cortex and striatum of the R/2 mouse model of Huntington's disease and subsequently improve motor symptoms. Thus, the in-vivo pharmacology profile of GLYN122 is a potential new preclinical candidate for the treatment of HD.
Collapse
Affiliation(s)
| | - Collin Y. Ewald
- grid.5801.c0000 0001 2156 2780Laboratory of Extracellular Matrix Regeneration, Department of Health Sciences and Technology, Institute of Translational Medicine, ETH Zürich, 8603 Schwerzenbach, Switzerland
| | - Xavier Jalencas
- grid.5841.80000 0004 1937 0247Chemotargets SL, Parc Científic de Barcelona, 08028 Barcelona, Catalonia Spain ,IMIM Hospital del Mar Medical Research Institute, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Catalonia Spain
| | - Shyam Masrani
- Medicxi Ventures, 25 Great Pulteney St, London, W1F 9NH UK
| | - Selin Meral
- Biomedical Center Munich of the University of Munich, Großhaderner Str. 9, 82152 Planegg, Germany
| | - Jordi Mestres
- grid.5841.80000 0004 1937 0247Chemotargets SL, Parc Científic de Barcelona, 08028 Barcelona, Catalonia Spain ,IMIM Hospital del Mar Medical Research Institute, Parc de Recerca Biomèdica de Barcelona (PRBB), 08003 Barcelona, Catalonia Spain
| |
Collapse
|
11
|
Fodale V, Pintauro R, Daldin M, Spiezia MC, Macdonald D, Bresciani A. Quantifying Huntingtin Protein in Human Cerebrospinal Fluid Using a Novel Polyglutamine Length-Independent Assay. J Huntingtons Dis 2022; 11:291-305. [PMID: 35938256 PMCID: PMC9535588 DOI: 10.3233/jhd-220527] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/02/2022]
Abstract
Background: The use of biomarkers has become a major component of clinical trial design. In Huntington’s disease (HD), quantifying the amount of huntingtin protein (HTT) in patient cerebrospinal fluid (CSF) has served as a pharmacodynamic readout for HTT-lowering therapeutic approaches and is a potential disease progression biomarker. To date, an ultrasensitive immunoassay to quantify mutant HTT protein (mHTT) has been used, but additional assays are needed to measure other forms of HTT protein. Objective: We aimed to develop an ultrasensitive immunoassay to quantify HTT protein in a polyglutamine length-independent manner (mHTT and non-expanded wild type HTT combined) in control and HD participant CSF samples. Methods: An ultrasensitive, bead-based, single molecule counting (SMC) immunoassay platform was used for the detection of HTT protein in human CSF samples. Results: A novel ultrasensitive SMC immunoassay was developed to quantify HTT protein in a polyglutamine length-independent manner and shown to measure HTT in both control and HD participant CSF samples. We validate the selectivity and specificity of the readout using biochemical and molecular biology tools, and we undertook a preliminary analytical qualification of this assay to enable its clinical use. We also used this novel assay, along with the previously described mHTT assay, to analyze CSF from control and HD participants. The results of this preliminary set suggests that correlation is present between mHTT and the polyglutamine length-independent HTT levels in human CSF. Conclusion: We have developed a novel ultrasensitive immunoassay that is able to quantify HTT protein in a polyglutamine length-independent manner in control and HD participant CSF.
Collapse
Affiliation(s)
- Valentina Fodale
- Department of Translational and Discovery Research, IRBM S.p.A., Pomezia, Roma, Italy
| | - Roberta Pintauro
- Department of Translational and Discovery Research, IRBM S.p.A., Pomezia, Roma, Italy
| | - Manuel Daldin
- Department of Translational and Discovery Research, IRBM S.p.A., Pomezia, Roma, Italy
| | | | | | - Alberto Bresciani
- Department of Translational and Discovery Research, IRBM S.p.A., Pomezia, Roma, Italy
| |
Collapse
|
12
|
Babi M, Neuman K, Peng CY, Maiuri T, Suart CE, Truant R. Recent Microscopy Advances and the Applications to Huntington’s Disease Research. J Huntingtons Dis 2022; 11:269-280. [PMID: 35848031 PMCID: PMC9484089 DOI: 10.3233/jhd-220536] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
Huntingtin is a 3144 amino acid protein defined as a scaffold protein with many intracellular locations that suggest functions in these compartments. Expansion of the CAG DNA tract in the huntingtin first exon is the cause of Huntington’s disease. An important tool in understanding the biological functions of huntingtin is molecular imaging at the single-cell level by microscopy and nanoscopy. The evolution of these technologies has accelerated since the Nobel Prize in Chemistry was awarded in 2014 for super-resolution nanoscopy. We are in a new era of light imaging at the single-cell level, not just for protein location, but also for protein conformation and biochemical function. Large-scale microscopy-based screening is also being accelerated by a coincident development of machine-based learning that offers a framework for truly unbiased data acquisition and analysis at very large scales. This review will summarize the newest technologies in light, electron, and atomic force microscopy in the context of unique challenges with huntingtin cell biology and biochemistry.
Collapse
Affiliation(s)
- Mouhanad Babi
- McMaster Centre for Advanced Light Microscopy (CALM) McMaster University, Hamilton, Canada
| | - Kaitlyn Neuman
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Christina Y. Peng
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Tamara Maiuri
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Celeste E. Suart
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
| | - Ray Truant
- Department of Biochemistry and Biomedical Sciences, McMaster University, Hamilton, Canada
- McMaster Centre for Advanced Light Microscopy (CALM) McMaster University, Hamilton, Canada
| |
Collapse
|
13
|
Tabrizi SJ, Estevez-Fraga C, van Roon-Mom WMC, Flower MD, Scahill RI, Wild EJ, Muñoz-Sanjuan I, Sampaio C, Rosser AE, Leavitt BR. Potential disease-modifying therapies for Huntington's disease: lessons learned and future opportunities. Lancet Neurol 2022; 21:645-658. [PMID: 35716694 PMCID: PMC7613206 DOI: 10.1016/s1474-4422(22)00121-1] [Citation(s) in RCA: 116] [Impact Index Per Article: 58.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2021] [Revised: 02/18/2022] [Accepted: 03/04/2022] [Indexed: 01/03/2023]
Abstract
Huntington's disease is the most frequent autosomal dominant neurodegenerative disorder; however, no disease-modifying interventions are available for patients with this disease. The molecular pathogenesis of Huntington's disease is complex, with toxicity that arises from full-length expanded huntingtin and N-terminal fragments of huntingtin, which are both prone to misfolding due to proteolysis; aberrant intron-1 splicing of the HTT gene; and somatic expansion of the CAG repeat in the HTT gene. Potential interventions for Huntington's disease include therapies targeting huntingtin DNA and RNA, clearance of huntingtin protein, DNA repair pathways, and other treatment strategies targeting inflammation and cell replacement. The early termination of trials of the antisense oligonucleotide tominersen suggest that it is time to reflect on lessons learned, where the field stands now, and the challenges and opportunities for the future.
Collapse
Affiliation(s)
- Sarah J Tabrizi
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK.
| | - Carlos Estevez-Fraga
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - Michael D Flower
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Rachael I Scahill
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | - Edward J Wild
- Huntington's Disease Centre, UCL Queen Square Institute of Neurology, University College London, London, UK
| | | | - Cristina Sampaio
- CHDI Management, CHDI Foundation Los Angeles, CA, USA; Laboratory of Clinical Pharmacology, Faculdade de Medicina de Lisboa, Lisbon, Portugal
| | - Anne E Rosser
- BRAIN unit, Division of Psychological Medicine and Clinical Neurosciences, School of Medicine, Cardiff University, Cardiff, UK
| | - Blair R Leavitt
- Centre for Huntington's disease, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
14
|
Ramírez-Jarquín UN, Sharma M, Zhou W, Shahani N, Subramaniam S. Deletion of SUMO1 attenuates behavioral and anatomical deficits by regulating autophagic activities in Huntington disease. Proc Natl Acad Sci U S A 2022; 119:e2107187119. [PMID: 35086928 PMCID: PMC8812691 DOI: 10.1073/pnas.2107187119] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2021] [Accepted: 12/13/2021] [Indexed: 01/18/2023] Open
Abstract
The CAG expansion of huntingtin (mHTT) associated with Huntington disease (HD) is a ubiquitously expressed gene, yet it prominently damages the striatum and cortex, followed by widespread peripheral defects as the disease progresses. However, the underlying mechanisms of neuronal vulnerability are unclear. Previous studies have shown that SUMO1 (small ubiquitin-like modifier-1) modification of mHtt promotes cellular toxicity, but the in vivo role and functions of SUMO1 in HD pathogenesis are unclear. Here, we report that SUMO1 deletion in Q175DN HD-het knockin mice (HD mice) prevented age-dependent HD-like motor and neurological impairments and suppressed the striatal atrophy and inflammatory response. SUMO1 deletion caused a drastic reduction in soluble mHtt levels and nuclear and extracellular mHtt inclusions while increasing cytoplasmic mHtt inclusions in the striatum of HD mice. SUMO1 deletion promoted autophagic activity, characterized by augmented interactions between mHtt inclusions and a lysosomal marker (LAMP1), increased LC3B- and LAMP1 interaction, and decreased interaction of sequestosome-1 (p62) and LAMP1 in DARPP-32-positive medium spiny neurons in HD mice. Depletion of SUMO1 in an HD cell model also diminished the mHtt levels and enhanced autophagy flux. In addition, the SUMOylation inhibitor ginkgolic acid strongly enhanced autophagy and diminished mHTT levels in human HD fibroblasts. These results indicate that SUMO is a critical therapeutic target in HD and that blocking SUMO may ameliorate HD pathogenesis by regulating autophagy activities.
Collapse
Affiliation(s)
| | - Manish Sharma
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458
| | - Wuyue Zhou
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458
| | - Neelam Shahani
- Department of Neuroscience, The Scripps Research Institute, Jupiter, FL 33458
| | | |
Collapse
|