1
|
Tesla R, Guhl C, Werthmann GC, Dixon D, Cenik B, Addepalli Y, Liang J, Fass DM, Rosenthal Z, Haggarty SJ, Williams NS, Posner BA, Ready JM, Herz J. Benzoxazole-derivatives enhance progranulin expression and reverse the aberrant lysosomal proteome caused by GRN haploinsufficiency. Nat Commun 2024; 15:6125. [PMID: 39033178 PMCID: PMC11271458 DOI: 10.1038/s41467-024-50076-8] [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: 05/29/2023] [Accepted: 07/01/2024] [Indexed: 07/23/2024] Open
Abstract
Heterozygous loss-of-function mutations in the GRN gene are a major cause of hereditary frontotemporal dementia. The mechanisms linking frontotemporal dementia pathogenesis to progranulin deficiency are not well understood, and there is currently no treatment. Our strategy to prevent the onset and progression of frontotemporal dementia in patients with GRN mutations is to utilize small molecule positive regulators of GRN expression to boost progranulin levels from the remaining functional GRN allele, thus restoring progranulin levels back to normal within the brain. This work describes a series of blood-brain-barrier-penetrant small molecules which significantly increase progranulin protein levels in human cellular models, correct progranulin protein deficiency in Grn+/- mouse brains, and reverse lysosomal proteome aberrations, a phenotypic hallmark of frontotemporal dementia, more efficiently than the previously described small molecule suberoylanilide hydroxamic acid. These molecules will allow further elucidation of the cellular functions of progranulin and its role in frontotemporal dementia and will also serve as lead structures for further drug development.
Collapse
Affiliation(s)
- Rachel Tesla
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for Translational Neurodegeneration Research, Dallas, TX, USA
| | - Charlotte Guhl
- Faculty of Chemistry and Earth Sciences, Institute of Organic Chemistry and Macromolecular Chemistry, Friedrich Schiller University Jena, 07743, Jena, Germany
| | - Gordon C Werthmann
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for Translational Neurodegeneration Research, Dallas, TX, USA
| | - Danielle Dixon
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for Translational Neurodegeneration Research, Dallas, TX, USA
| | - Basar Cenik
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for Translational Neurodegeneration Research, Dallas, TX, USA
| | - Yesu Addepalli
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jue Liang
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Daniel M Fass
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Zachary Rosenthal
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Massachusetts General Hospital, Boston, MA, USA
- Department of Chemistry & Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Stephen J Haggarty
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Massachusetts General Hospital, Harvard Medical School, Boston, MA, USA
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Bruce A Posner
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joseph M Ready
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Center for Translational Neurodegeneration Research, Dallas, TX, USA.
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
2
|
Pasternak M, Mirza SS, Luciw N, Mutsaerts HJMM, Petr J, Thomas D, Cash D, Bocchetta M, Tartaglia MC, Mitchell SB, Black SE, Freedman M, Tang‐Wai D, Rogaeva E, Russell LL, Bouzigues A, van Swieten JC, Jiskoot LC, Seelaar H, Laforce R, Tiraboschi P, Borroni B, Galimberti D, Rowe JB, Graff C, Finger E, Sorbi S, de Mendonça A, Butler C, Gerhard A, Sanchez‐Valle R, Moreno F, Synofzik M, Vandenberghe R, Ducharme S, Levin J, Otto M, Santana I, Strafella AP, MacIntosh BJ, Rohrer JD, Masellis M. Longitudinal cerebral perfusion in presymptomatic genetic frontotemporal dementia: GENFI results. Alzheimers Dement 2024; 20:3525-3542. [PMID: 38623902 PMCID: PMC11095434 DOI: 10.1002/alz.13750] [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: 09/21/2023] [Revised: 01/16/2024] [Accepted: 01/21/2024] [Indexed: 04/17/2024]
Abstract
INTRODUCTION Effective longitudinal biomarkers that track disease progression are needed to characterize the presymptomatic phase of genetic frontotemporal dementia (FTD). We investigate the utility of cerebral perfusion as one such biomarker in presymptomatic FTD mutation carriers. METHODS We investigated longitudinal profiles of cerebral perfusion using arterial spin labeling magnetic resonance imaging in 42 C9orf72, 70 GRN, and 31 MAPT presymptomatic carriers and 158 non-carrier controls. Linear mixed effects models assessed perfusion up to 5 years after baseline assessment. RESULTS Perfusion decline was evident in all three presymptomatic groups in global gray matter. Each group also featured its own regional pattern of hypoperfusion over time, with the left thalamus common to all groups. Frontal lobe regions featured lower perfusion in those who symptomatically converted versus asymptomatic carriers past their expected age of disease onset. DISCUSSION Cerebral perfusion is a potential biomarker for assessing genetic FTD and its genetic subgroups prior to symptom onset. HIGHLIGHTS Gray matter perfusion declines in at-risk genetic frontotemporal dementia (FTD). Regional perfusion decline differs between at-risk genetic FTD subgroups . Hypoperfusion in the left thalamus is common across all presymptomatic groups. Converters exhibit greater right frontal hypoperfusion than non-converters past their expected conversion date. Cerebral hypoperfusion is a potential early biomarker of genetic FTD.
Collapse
|
3
|
Rosenthal ZC, Fass DM, Payne NC, She A, Patnaik D, Hennig KM, Tesla R, Werthmann GC, Guhl C, Reis SA, Wang X, Chen Y, Placzek M, Williams NS, Hooker J, Herz J, Mazitschek R, Haggarty SJ. Epigenetic modulation through BET bromodomain inhibitors as a novel therapeutic strategy for progranulin-deficient frontotemporal dementia. Sci Rep 2024; 14:9064. [PMID: 38643236 PMCID: PMC11032351 DOI: 10.1038/s41598-024-59110-7] [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: 12/06/2023] [Accepted: 04/08/2024] [Indexed: 04/22/2024] Open
Abstract
Frontotemporal dementia (FTD) is a debilitating neurodegenerative disorder with currently no disease-modifying treatment options available. Mutations in GRN are one of the most common genetic causes of FTD, near ubiquitously resulting in progranulin (PGRN) haploinsufficiency. Small molecules that can restore PGRN protein to healthy levels in individuals bearing a heterozygous GRN mutation may thus have therapeutic value. Here, we show that epigenetic modulation through bromodomain and extra-terminal domain (BET) inhibitors (BETi) potently enhance PGRN protein levels, both intracellularly and secreted forms, in human central nervous system (CNS)-relevant cell types, including in microglia-like cells. In terms of potential for disease modification, we show BETi treatment effectively restores PGRN levels in neural cells with a GRN mutation known to cause PGRN haploinsufficiency and FTD. We demonstrate that BETi can rapidly and durably enhance PGRN in neural progenitor cells (NPCs) in a manner dependent upon BET protein expression, suggesting a gain-of-function mechanism. We further describe a CNS-optimized BETi chemotype that potently engages endogenous BRD4 and enhances PGRN expression in neuronal cells. Our results reveal a new epigenetic target for treating PGRN-deficient forms of FTD and provide mechanistic insight to aid in translating this discovery into therapeutics.
Collapse
Affiliation(s)
- Zachary C Rosenthal
- Chemical Neurobiology Laboratory, Precision Therapeutics Unit, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
- Department of Chemistry & Chemical Biology, Harvard University, Cambridge, MA, USA
| | - Daniel M Fass
- Chemical Neurobiology Laboratory, Precision Therapeutics Unit, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - N Connor Payne
- Department of Chemistry & Chemical Biology, Harvard University, Cambridge, MA, USA
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
| | - Angela She
- Chemical Neurobiology Laboratory, Precision Therapeutics Unit, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Debasis Patnaik
- Chemical Neurobiology Laboratory, Precision Therapeutics Unit, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Krista M Hennig
- Chemical Neurobiology Laboratory, Precision Therapeutics Unit, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Rachel Tesla
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Gordon C Werthmann
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Charlotte Guhl
- Faculty of Chemistry and Earth Sciences, Institute of Organic Chemistry and Macromolecular Chemistry, Friedrich Schiller University Jena, Jena, Germany
| | - Surya A Reis
- Chemical Neurobiology Laboratory, Precision Therapeutics Unit, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA
| | - Xiaoyu Wang
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Yueting Chen
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Michael Placzek
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Noelle S Williams
- Department of Biochemistry, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jacob Hooker
- Department of Radiology, Harvard Medical School, Boston, MA, USA
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital, Boston, MA, USA
| | - Joachim Herz
- Department of Molecular Genetics, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Neuroscience, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Neurology and Neurotherapeutics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Ralph Mazitschek
- Center for Systems Biology, Massachusetts General Hospital, Boston, MA, USA
- Harvard T.H. Chan School of Public Health, Boston, MA, USA
- Broad Institute of MIT and Harvard, Cambridge, MA, USA
| | - Stephen J Haggarty
- Chemical Neurobiology Laboratory, Precision Therapeutics Unit, Center for Genomic Medicine, Departments of Neurology and Psychiatry, Harvard Medical School, Massachusetts General Hospital, Boston, MA, USA.
| |
Collapse
|
4
|
Kertesz A, Finger E, Munoz DG. Progress in Primary Progressive Aphasia: A Review. Cogn Behav Neurol 2024; 37:3-12. [PMID: 38498721 DOI: 10.1097/wnn.0000000000000365] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2023] [Accepted: 08/07/2023] [Indexed: 03/20/2024]
Abstract
We present a review of the definition, classification, and epidemiology of primary progressive aphasia (PPA); an update of the taxonomy of the clinical syndrome of PPA; and recent advances in the neuroanatomy, pathology, and genetics of PPA, as well as the search for biomarkers and treatment. PPA studies that have contributed to concepts of language organization and disease propagation in neurodegeneration are also reviewed. In addition, the issues of heterogeneity versus the relationships of the clinical phenotypes and their relationship to biological, pathological, and genetic advances are discussed, as is PPA's relationship to other conditions such as frontotemporal dementia, corticobasal degeneration, progressive supranuclear palsy, Pick disease, and amyotrophic lateral sclerosis. Arguments are presented in favor of considering these conditions as one entity versus many.
Collapse
Affiliation(s)
- Andrew Kertesz
- Department of Clinical Neurosciences, Western University Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | - Elizabeth Finger
- Department of Clinical Neurosciences, Western University Schulich School of Medicine & Dentistry, London, Ontario, Canada
| | - David G Munoz
- Department of Pathology, St Michael's Hospital, Toronto, Ontario, Canada
| |
Collapse
|
5
|
Ducharme S, Pijnenburg Y, Rohrer JD, Huey E, Finger E, Tatton N. Identifying and Diagnosing TDP-43 Neurodegenerative Diseases in Psychiatry. Am J Geriatr Psychiatry 2024; 32:98-113. [PMID: 37741764 PMCID: PMC11270911 DOI: 10.1016/j.jagp.2023.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/02/2023] [Accepted: 08/24/2023] [Indexed: 09/25/2023]
Abstract
Neuropsychiatric symptoms (NPS) are common manifestations of neurodegenerative disorders and are often early signs of those diseases. Among those neurodegenerative diseases, TDP-43 proteinopathies are an increasingly recognized cause of early neuropsychiatric manifestations. TDP-43-related diseases include frontotemporal dementia (FTD), amyotrophic lateral sclerosis (ALS), and Limbic-Predominant Age-Related TDP-43 Encephalopathy (LATE). The majority of TDP-43-related diseases are sporadic, but a significant proportion is hereditary, with progranulin (GRN) mutations and C9orf72 repeat expansions as the most common genetic etiologies. Studies reveal that NPS can be the initial manifestation of those diseases or can complicate disease course, but there is a lack of awareness among clinicians about TDP-43-related diseases, which leads to common diagnostic mistakes or delays. There is also emerging evidence that TDP-43 accumulations could play a role in late-onset primary psychiatric disorders. In the absence of robust biomarkers for TDP-43, the diagnosis remains primarily based on clinical assessment and neuroimaging. Given the association with psychiatric symptoms, clinical psychiatrists have a key role in the early identification of patients with TDP-43-related diseases. This narrative review provides a comprehensive overview of the pathobiology of TDP-43, resulting clinical presentations, and associated neuropsychiatric manifestations to help guide clinical practice.
Collapse
Affiliation(s)
- Simon Ducharme
- Department of Psychiatry (SD), Douglas Mental Health University Institute, McGill University, Montreal, Canada; McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Canada.
| | - Yolande Pijnenburg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience (YP), Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease (JDR), UCL Queen Square Institute of Neurology, London, UK
| | - Edward Huey
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Department of Psychiatry (EH), Columbia University, New York, NY
| | - Elizabeth Finger
- London Health Sciences Centre Parkwood Institute (EF), London, ON, Canada
| | | |
Collapse
|
6
|
Aggarwal G, Banerjee S, Jones SA, Benchaar Y, Bélanger J, Sévigny M, Smith DM, Niehoff ML, Pavlack M, de Vera IMS, Petkau TL, Leavitt BR, Ling K, Jafar-Nejad P, Rigo F, Morley JE, Farr SA, Dutchak PA, Sephton CF, Nguyen AD. Antisense oligonucleotides targeting the miR-29b binding site in the GRN mRNA increase progranulin translation. J Biol Chem 2023; 299:105475. [PMID: 37981208 PMCID: PMC10755782 DOI: 10.1016/j.jbc.2023.105475] [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: 09/18/2023] [Revised: 10/13/2023] [Accepted: 11/07/2023] [Indexed: 11/21/2023] Open
Abstract
Heterozygous GRN (progranulin) mutations cause frontotemporal dementia (FTD) due to haploinsufficiency, and increasing progranulin levels is a major therapeutic goal. Several microRNAs, including miR-29b, negatively regulate progranulin protein levels. Antisense oligonucleotides (ASOs) are emerging as a promising therapeutic modality for neurological diseases, but strategies for increasing target protein levels are limited. Here, we tested the efficacy of ASOs as enhancers of progranulin expression by sterically blocking the miR-29b binding site in the 3' UTR of the human GRN mRNA. We found 16 ASOs that increase progranulin protein in a dose-dependent manner in neuroglioma cells. A subset of these ASOs also increased progranulin protein in iPSC-derived neurons and in a humanized GRN mouse model. In FRET-based assays, the ASOs effectively competed for miR-29b from binding to the GRN 3' UTR RNA. The ASOs increased levels of newly synthesized progranulin protein by increasing its translation, as revealed by polysome profiling. Together, our results demonstrate that ASOs can be used to effectively increase target protein levels by partially blocking miR binding sites. This ASO strategy may be therapeutically feasible for progranulin-deficient FTD as well as other conditions of haploinsufficiency.
Collapse
Affiliation(s)
- Geetika Aggarwal
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA; Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St Louis, Missouri, USA; Institute for Translational Neuroscience, Saint Louis University, St Louis, Missouri, USA
| | - Subhashis Banerjee
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA; Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St Louis, Missouri, USA; Institute for Translational Neuroscience, Saint Louis University, St Louis, Missouri, USA
| | - Spencer A Jones
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA; Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St Louis, Missouri, USA; Institute for Translational Neuroscience, Saint Louis University, St Louis, Missouri, USA
| | - Yousri Benchaar
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Jasmine Bélanger
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Myriam Sévigny
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Denise M Smith
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA; Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St Louis, Missouri, USA; Institute for Translational Neuroscience, Saint Louis University, St Louis, Missouri, USA
| | - Michael L Niehoff
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA; Veterans Affairs Medical Center, St Louis, Missouri, USA
| | - Monica Pavlack
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St Louis, Missouri, USA; Institute for Translational Neuroscience, Saint Louis University, St Louis, Missouri, USA
| | - Ian Mitchelle S de Vera
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St Louis, Missouri, USA; Institute for Translational Neuroscience, Saint Louis University, St Louis, Missouri, USA
| | - Terri L Petkau
- Department of Medical Genetics, Centre for Molecular Medicine & Therapeutics, B.C. Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada
| | - Blair R Leavitt
- Department of Medical Genetics, Centre for Molecular Medicine & Therapeutics, B.C. Children's Hospital, University of British Columbia, Vancouver, British Columbia, Canada; Division of Neurology, Department of Medicine, University of British Columbia Hospital, Vancouver, British Columbia, Canada; Center for Brain Health, University of British Columbia, Vancouver, British Columbia, Canada
| | - Karen Ling
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | | | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, California, USA
| | - John E Morley
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA
| | - Susan A Farr
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA; Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St Louis, Missouri, USA; Institute for Translational Neuroscience, Saint Louis University, St Louis, Missouri, USA; Veterans Affairs Medical Center, St Louis, Missouri, USA
| | - Paul A Dutchak
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Chantelle F Sephton
- Department of Psychiatry and Neuroscience, CERVO Brain Research Centre, Laval University, Quebec City, Quebec, Canada
| | - Andrew D Nguyen
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St Louis, Missouri, USA; Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St Louis, Missouri, USA; Institute for Translational Neuroscience, Saint Louis University, St Louis, Missouri, USA.
| |
Collapse
|
7
|
Mukherjee A, Zamani F, Suzuki T. Evolution of Slow-Binding Inhibitors Targeting Histone Deacetylase Isoforms. J Med Chem 2023; 66:11672-11700. [PMID: 37651268 DOI: 10.1021/acs.jmedchem.3c01160] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/02/2023]
Abstract
Because the overexpression of histone deacetylase enzymes (HDACs) has been linked to numerous diseases, including various cancers and neurodegenerative disorders, HDAC inhibitors have emerged as promising therapeutic agents. However, most HDAC inhibitors lack both subclass and isoform selectivity, which leads to potential toxicity. Unlike classical hydroxamate HDAC inhibitors, slow-binding HDAC inhibitors form tight and prolonged bonds with HDAC enzymes. This distinct mechanism of action improves both selectivity and toxicity profiles, which makes slow-binding HDAC inhibitors a promising class of therapeutic agents for various diseases. Therefore, the development of slow-binding HDAC inhibitors that can effectively target a wide range of HDAC isoforms is crucial. This Perspective provides valuable insights into the potential and progress of slow-binding HDAC inhibitors as promising drug candidates for the treatment of various diseases.
Collapse
Affiliation(s)
| | - Farzad Zamani
- SANKEN, Osaka University, Mihogaoka, Ibaraki, Osaka 567-0047, Japan
| | - Takayoshi Suzuki
- SANKEN, Osaka University, Mihogaoka, Ibaraki, Osaka 567-0047, Japan
| |
Collapse
|
8
|
Smith DM, Niehoff ML, Ling K, Jafar-Nejad P, Rigo F, Farr SA, Wilkinson MF, Nguyen AD. Targeting nonsense-mediated RNA decay does not increase progranulin levels in the Grn R493X mouse model of frontotemporal dementia. PLoS One 2023; 18:e0282822. [PMID: 36893203 PMCID: PMC9997918 DOI: 10.1371/journal.pone.0282822] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2022] [Accepted: 02/23/2023] [Indexed: 03/10/2023] Open
Abstract
A common cause of frontotemporal dementia (FTD) are nonsense mutations in the progranulin (GRN) gene. Because nonsense mutations activate the nonsense-mediated RNA decay (NMD) pathway, we sought to inhibit this RNA turnover pathway as a means to increase progranulin levels. Using a knock-in mouse model harboring a common patient mutation, we tested whether either pharmacological or genetic inhibition of NMD upregulates progranulin in these GrnR493X mice. We first examined antisense oligonucleotides (ASOs) targeting an exonic region in GrnR493X mRNA predicted to block its degradation by NMD. As we previously reported, these ASOs effectively increased GrnR493X mRNA levels in fibroblasts in vitro. However, following CNS delivery, we found that none of the 8 ASOs we tested increased Grn mRNA levels in the brains of GrnR493X mice. This result was obtained despite broad ASO distribution in the brain. An ASO targeting a different mRNA was effective when administered in parallel to wild-type mice. As an independent approach to inhibit NMD, we examined the effect of loss of an NMD factor not required for embryonic viability: UPF3b. We found that while Upf3b deletion effectively perturbed NMD, it did not increase Grn mRNA levels in Grn+/R493X mouse brains. Together, our results suggest that the NMD-inhibition approaches that we used are likely not viable for increasing progranulin levels in individuals with FTD caused by nonsense GRN mutations. Thus, alternative approaches should be pursued.
Collapse
Affiliation(s)
- Denise M. Smith
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
- Institute for Translational Neuroscience, Saint Louis University, St. Louis, Missouri, United States of America
| | - Michael L. Niehoff
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
- Veterans Affairs Medical Center, St. Louis, Missouri, United States of America
| | - Karen Ling
- Ionis Pharmaceuticals, Carlsbad, California, United States of America
| | | | - Frank Rigo
- Ionis Pharmaceuticals, Carlsbad, California, United States of America
| | - Susan A. Farr
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
- Institute for Translational Neuroscience, Saint Louis University, St. Louis, Missouri, United States of America
- Veterans Affairs Medical Center, St. Louis, Missouri, United States of America
| | - Miles F. Wilkinson
- Department of Obstetrics, Gynecology, and Reproductive Sciences, University of California San Diego, La Jolla, California, United States of America
- Institute of Genomic Medicine, University of California San Diego, La Jolla, California, United States of America
| | - Andrew D. Nguyen
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
- Department of Pharmacology and Physiology, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
- Institute for Translational Neuroscience, Saint Louis University, St. Louis, Missouri, United States of America
| |
Collapse
|
9
|
Jiang D, Li T, Guo C, Tang TS, Liu H. Small molecule modulators of chromatin remodeling: from neurodevelopment to neurodegeneration. Cell Biosci 2023; 13:10. [PMID: 36647159 PMCID: PMC9841685 DOI: 10.1186/s13578-023-00953-4] [Citation(s) in RCA: 21] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2022] [Accepted: 01/03/2023] [Indexed: 01/18/2023] Open
Abstract
The dynamic changes in chromatin conformation alter the organization and structure of the genome and further regulate gene transcription. Basically, the chromatin structure is controlled by reversible, enzyme-catalyzed covalent modifications to chromatin components and by noncovalent ATP-dependent modifications via chromatin remodeling complexes, including switch/sucrose nonfermentable (SWI/SNF), inositol-requiring 80 (INO80), imitation switch (ISWI) and chromodomain-helicase DNA-binding protein (CHD) complexes. Recent studies have shown that chromatin remodeling is essential in different stages of postnatal and adult neurogenesis. Chromatin deregulation, which leads to defects in epigenetic gene regulation and further pathological gene expression programs, often causes a wide range of pathologies. This review first gives an overview of the regulatory mechanisms of chromatin remodeling. We then focus mainly on discussing the physiological functions of chromatin remodeling, particularly histone and DNA modifications and the four classes of ATP-dependent chromatin-remodeling enzymes, in the central and peripheral nervous systems under healthy and pathological conditions, that is, in neurodegenerative disorders. Finally, we provide an update on the development of potent and selective small molecule modulators targeting various chromatin-modifying proteins commonly associated with neurodegenerative diseases and their potential clinical applications.
Collapse
Affiliation(s)
- Dongfang Jiang
- grid.458458.00000 0004 1792 6416State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China ,grid.410726.60000 0004 1797 8419Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, 100101 China
| | - Tingting Li
- grid.458458.00000 0004 1792 6416State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China ,grid.410726.60000 0004 1797 8419Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, 100101 China
| | - Caixia Guo
- grid.9227.e0000000119573309Beijing Institute of Genomics, Chinese Academy of Sciences/China National Center for Bioinformation, Beijing, 100101 China ,grid.410726.60000 0004 1797 8419Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, 100101 China
| | - Tie-Shan Tang
- grid.458458.00000 0004 1792 6416State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China ,grid.512959.3Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101 China ,grid.410726.60000 0004 1797 8419Chinese Academy of Sciences, University of Chinese Academy of Sciences, Beijing, 100101 China
| | - Hongmei Liu
- grid.458458.00000 0004 1792 6416State Key Laboratory of Membrane Biology, Institute of Zoology, Chinese Academy of Sciences, Beijing, 100101 China ,grid.512959.3Beijing Institute for Stem Cell and Regenerative Medicine, Beijing, 100101 China
| |
Collapse
|
10
|
Kashyap SN, Boyle NR, Roberson ED. Preclinical Interventions in Mouse Models of Frontotemporal Dementia Due to Progranulin Mutations. Neurotherapeutics 2023; 20:140-153. [PMID: 36781744 PMCID: PMC10119358 DOI: 10.1007/s13311-023-01348-6] [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] [Accepted: 01/20/2023] [Indexed: 02/15/2023] Open
Abstract
Heterozygous loss-of-function mutations in progranulin (GRN) cause frontotemporal dementia (FTD), a leading cause of early-onset dementia characterized clinically by behavioral, social, and language deficits. There are currently no FDA-approved therapeutics for FTD-GRN, but this has been an active area of investigation, and several approaches are now in clinical trials. Here, we review preclinical development of therapies for FTD-GRN with a focus on testing in mouse models. Since most FTD-GRN-associated mutations cause progranulin haploinsufficiency, these approaches focus on raising progranulin levels. We begin by considering the disorders associated with altered progranulin levels, and then review the basics of progranulin biology including its lysosomal, neurotrophic, and immunomodulatory functions. We discuss mouse models of progranulin insufficiency and how they have been used in preclinical studies on a variety of therapeutic approaches. These include approaches to raise progranulin expression from the normal allele or facilitate progranulin production by the mutant allele, as well as approaches to directly increase progranulin levels by delivery across the blood-brain barrier or by gene therapy. Several of these approaches have entered clinical trials, providing hope that new therapies for FTD-GRN may be the next frontier in the treatment of neurodegenerative disease.
Collapse
Affiliation(s)
- Shreya N Kashyap
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Medical Scientist Training Program, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Nicholas R Boyle
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Medical Scientist Training Program, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Erik D Roberson
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Medical Scientist Training Program, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
11
|
Zhang L, Liu Y, Lu Y, Wang G. Targeting epigenetics as a promising therapeutic strategy for treatment of neurodegenerative diseases. Biochem Pharmacol 2022; 206:115295. [DOI: 10.1016/j.bcp.2022.115295] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2022] [Revised: 10/03/2022] [Accepted: 10/04/2022] [Indexed: 11/16/2022]
|
12
|
Recent Advances in Frontotemporal Dementia. Neurol Sci 2022:1-10. [DOI: 10.1017/cjn.2022.69] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
13
|
Rhinn H, Tatton N, McCaughey S, Kurnellas M, Rosenthal A. Progranulin as a therapeutic target in neurodegenerative diseases. Trends Pharmacol Sci 2022; 43:641-652. [PMID: 35039149 DOI: 10.1016/j.tips.2021.11.015] [Citation(s) in RCA: 98] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 11/12/2021] [Accepted: 11/18/2021] [Indexed: 01/02/2023]
Abstract
Progranulin (PGRN, encoded by the GRN gene) plays a key role in the development, survival, function, and maintenance of neurons and microglia in the mammalian brain. It regulates lysosomal biogenesis, inflammation, repair, stress response, and aging. GRN loss-of-function mutations cause neuronal ceroid lipofuscinosis or frontotemporal dementia-GRN (FTD-GRN) in a gene dosage-dependent manner. Mutations that reduce PGRN levels increase the risk for developing Alzheimer's disease, Parkinson's disease, and limbic-predominant age-related transactivation response DNA-binding protein 43 encephalopathy, as well as exacerbate the progression of amyotrophic lateral sclerosis (ALS) and FTD caused by the hexanucleotide repeat expansion in the C9orf72 gene. Elevating and/or restoring PGRN levels is an attractive therapeutic strategy and is being investigated for neurodegenerative diseases through multiple mechanisms of action.
Collapse
Affiliation(s)
- Herve Rhinn
- Alector, Inc., South San Francisco, CA 94080, USA
| | | | | | | | | |
Collapse
|
14
|
Rahman M, Mim SA, Islam R, Parvez A, Islam F, Uddin MB, Rahaman S, Shuvo PA, Ahmed M, Greig NH, Kamal MA. Exploring the Recent Trends in Management of Dementia and Frailty: Focus on Diagnosis and Treatment. Curr Med Chem 2022; 29:5289-5314. [PMID: 35400321 PMCID: PMC10477961 DOI: 10.2174/0929867329666220408102051] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2021] [Revised: 09/19/2021] [Accepted: 09/19/2021] [Indexed: 02/08/2023]
Abstract
Dementia and frailty increase health adversities in older adults, which are topics of growing research interest. Frailty is considered to correspond to a biological syndrome associated with age. Frail patients may ultimately develop multiple dysfunctions across several systems, including stroke, transient ischemic attack, vascular dementia, Parkinson's disease, Alzheimer's disease, frontotemporal dementia, dementia with Lewy bodies, cortico-basal degeneration, multiple system atrophy, amyotrophic lateral sclerosis, and Creutzfeldt-Jakob disease. Patients with dementia and frailty often develop malnutrition and weight loss. Rigorous nutritional, pharmacological, and non-pharmacological interventions generally are required for these patients, which is a challenging issue for healthcare providers. A healthy diet and lifestyle instigated at an early age can reduce the risk of frailty and dementia. For optimal treatment, accurate diagnosis involving clinical evaluation, cognitive screening, essential laboratory evaluation, structural imaging, functional neuroimaging, and neuropsychological testing is necessary. Diagnosis procedures best apply the clinical diagnosis, identifying the cause(s) and the condition(s) appropriate for treatment. The patient's history, caregiver's interview, physical examination, cognitive evaluation, laboratory tests, and structural imaging should best be involved in the diagnostic process. Varying types of physical exercise can aid the treatment of these disorders. Nutrition maintenance is a particularly significant factor, such as exceptionally high-calorie dietary supplements and a Mediterranean diet to support weight gain. The core purpose of this article is to investigate trends in the management of dementia and frailty, focusing on improving diagnosis and treatment. Substantial evidence builds the consensus that a combination of balanced nutrition and good physical activity is an integral part of treatment. Notably, more evidence-based medicine knowledge is required.
Collapse
Affiliation(s)
- Mominur Rahman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka-1207, Bangladesh
| | - Sadia Afsana Mim
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka-1207, Bangladesh
| | - Rezaul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka-1207, Bangladesh
| | - Anwar Parvez
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka-1207, Bangladesh
| | - Fahadul Islam
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka-1207, Bangladesh
| | - Mohammad Borhan Uddin
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka-1207, Bangladesh
| | - Saidur Rahaman
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka-1207, Bangladesh
| | - Pollob Ahmed Shuvo
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka-1207, Bangladesh
| | - Muniruddin Ahmed
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka-1207, Bangladesh
| | - Nigel H. Greig
- Translational Gerontology Branch, Intramural Research Program, National Institute on Aging, National Institutes of Health, Baltimore, MD 21224, USA
| | - Mohammad Amjad Kamal
- Department of Pharmacy, Faculty of Allied Health Sciences, Daffodil International University, Dhaka-1207, Bangladesh
- Institutes for Systems Genetics, Frontiers Science Center for Disease-related Molecular Network, West China Hospital, Sichuan University, Chengdu 610041, Sichuan, China
- King Fahd Medical Research Center, King Abdulaziz University, Saudi Arabia
- Enzymoics, NSW; Novel Global Community Educational Foundation, Peterlee Place, Hebersham, NSW 2770, Australia
| |
Collapse
|
15
|
Abstract
Neuroepigenetics, a new branch of epigenetics, plays an important role in the regulation of gene expression. Neuroepigenetics is associated with holistic neuronal function and helps in formation and maintenance of memory and learning processes. This includes neurodevelopment and neurodegenerative defects in which histone modification enzymes appear to play a crucial role. These modifications, carried out by acetyltransferases and deacetylases, regulate biologic and cellular processes such as apoptosis and autophagy, inflammatory response, mitochondrial dysfunction, cell-cycle progression and oxidative stress. Alterations in acetylation status of histone as well as non-histone substrates lead to transcriptional deregulation. Histone deacetylase decreases acetylation status and causes transcriptional repression of regulatory genes involved in neural plasticity, synaptogenesis, synaptic and neural plasticity, cognition and memory, and neural differentiation. Transcriptional deactivation in the brain results in development of neurodevelopmental and neurodegenerative disorders. Mounting evidence implicates histone deacetylase inhibitors as potential therapeutic targets to combat neurologic disorders. Recent studies have targeted naturally-occurring biomolecules and micro-RNAs to improve cognitive defects and memory. Multi-target drug ligands targeting HDAC have been developed and used in cell-culture and animal-models of neurologic disorders to ameliorate synaptic and cognitive dysfunction. Herein, we focus on the implications of histone deacetylase enzymes in neuropathology, their regulation of brain function and plausible involvement in the pathogenesis of neurologic defects.
Collapse
|
16
|
Ljubenkov PA, Edwards L, Iaccarino L, La Joie R, Rojas JC, Koestler M, Harris B, Boeve BF, Borroni B, van Swieten JC, Grossman M, Pasquier F, Frisoni GB, Mummery CJ, Vandenberghe R, Le Ber I, Hannequin D, McGinnis SM, Auriacombe S, Onofrj M, Goodman IJ, Riordan HJ, Wisniewski G, Hesterman J, Marek K, Haynes BA, Patzke H, Koenig G, Hilt D, Moebius H, Boxer AL. Effect of the Histone Deacetylase Inhibitor FRM-0334 on Progranulin Levels in Patients With Progranulin Gene Haploinsufficiency: A Randomized Clinical Trial. JAMA Netw Open 2021; 4:e2125584. [PMID: 34559230 PMCID: PMC8463943 DOI: 10.1001/jamanetworkopen.2021.25584] [Citation(s) in RCA: 17] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/14/2022] Open
Abstract
IMPORTANCE Histone deacetylase inhibitors have been repeatedly shown to elevate progranulin levels in preclinical models. This report describes the first randomized clinical trial of a histone deacetylase inhibitor in frontotemporal dementia (FTD) resulting from progranulin (GRN) gene variations. OBJECTIVE To characterize the safety, tolerability, plasma pharmacokinetics, and pharmacodynamic effects of oral FRM-0334 on plasma progranulin and other exploratory biomarkers, including fluorodeoxyglucose (FDG)-positron emission tomography (PET), in individuals with GRN haploinsufficiency. DESIGN, SETTING, AND PARTICIPANTS In this randomized, double-blind, placebo-controlled, dose-escalating, phase 2a safety, tolerability, and pharmacodynamic clinical study, 2 doses of a histone deacetylase inhibitor (FRM-0334) were administered to participants with prodromal to moderate FTD with granulin variations. Participants were recruited from January 13, 2015, to April 13, 2016. The study included 27 participants with prodromal (n = 8) or mild-to-moderate symptoms of FTD (n = 19) and heterozygous pathogenic variations in GRN and was conducted at multiple centers in North America, the UK, and the European Union. Data were analyzed from June 9, 2019, to May 13, 2021. INTERVENTIONS Daily oral placebo (n = 5), 300 mg of FRM-0334 (n = 11), or 500 mg of FRM-0334 (n = 11) was administered for 28 days. MAIN OUTCOMES AND MEASURES Primary outcomes were safety and tolerability of FRM-0334 and its peripheral pharmacodynamic effect on plasma progranulin. Secondary outcomes were the plasma pharmacokinetic profile of FRM-0334 and its pharmacodynamic effect on cerebrospinal fluid progranulin. Exploratory outcomes were FDG-PET, FTD clinical severity, and cerebrospinal fluid biomarkers (neurofilament light chain [NfL], amyloid β 1-42, phosphorylated tau 181, and total tau [t-tau]). RESULTS A total of 27 participants (mean [SD] age, 56.6 [10.5] years; 16 women [59.3%]; 26 White participants [96.3%]) with GRN variations were randomized and completed treatment. FRM-0334 was safe and well tolerated but did not affect plasma progranulin (4.3 pg/mL per day change after treatment; 95% CI, -10.1 to 18.8 pg/mL; P = .56), cerebrospinal fluid progranulin (0.42 pg/mL per day; 95% CI, -0.12 to 0.95 pg/mL; P = .13), or exploratory pharmacodynamic measures. Plasma FRM-0334 exposure did not increase proportionally with dose. Brain FDG-PET data were available in 26 of 27 randomized participants. In a cross-sectional analysis of 26 individuals, bifrontal cortical FDG hypometabolism was associated with worse Clinical Dementia Rating (CDR) plus National Alzheimer's Coordinating Center frontotemporal lobar degeneration sum of boxes score (b = -3.6 × 10-2 standardized uptake value ratio [SUVR] units/CDR units; 95% CI, -4.9 × 10-2 to -2.2 × 10-2; P < .001), high cerebrospinal fluid NfL (b = -9.2 × 10-5 SUVR units/pg NfL/mL; 95% CI, -1.3 × 10-4 to -5.6 × 10-5; P < .001), and high CSF t-tau (-7.2 × 10-4 SUVR units/pg t-tau/mL; 95% CI, -1.4 × 10-3 to -9.5 × 10-5; P = .03). CONCLUSIONS AND RELEVANCE In this randomized clinical trial, the current formulation of FRM-0334 did not elevate PRGN levels, which could reflect a lack of efficacy at attained exposures, low bioavailability, or some combination of the 2 factors. Bifrontal FDG-PET is a sensitive measure of symptomatic GRN haploinsufficiency. International multicenter clinical trials of FTD-GRN are feasible. TRIAL REGISTRATION ClinicalTrials.gov Identifier: NCT02149160.
Collapse
Affiliation(s)
- Peter A. Ljubenkov
- Memory and Aging Center, Department of Neurology, University of California at San Francisco, Weill Institute for Neurosciences, San Francisco
| | - Lauren Edwards
- Memory and Aging Center, Department of Neurology, University of California at San Francisco, Weill Institute for Neurosciences, San Francisco
| | - Leonardo Iaccarino
- Memory and Aging Center, Department of Neurology, University of California at San Francisco, Weill Institute for Neurosciences, San Francisco
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, University of California at San Francisco, Weill Institute for Neurosciences, San Francisco
| | - Julio C. Rojas
- Memory and Aging Center, Department of Neurology, University of California at San Francisco, Weill Institute for Neurosciences, San Francisco
| | - Mary Koestler
- Memory and Aging Center, Department of Neurology, University of California at San Francisco, Weill Institute for Neurosciences, San Francisco
| | - Baruch Harris
- ROME Therapeutics, Cambridge, Massachusetts
- Metera Pharmaceuticals, Cambridge, Massachusetts
| | | | - Barbara Borroni
- Neurology Unit, Centre for Neurodegenerative Disorders, ASST Spedali Civili Brescia, Italy
- Department of Clinical and Experimental Sciences, University of Brescia, Italy
| | - John C. van Swieten
- Department of Neurology, Erasmus University Medical Center, Rotterdam, the Netherlands
| | - Murray Grossman
- Perelman School of Medicine, University of Pennsylvania, Philadelphia
| | - Florence Pasquier
- Lille University, Inserm 1172, CHU Lille, CNR-MAJ, DISTALZ, LiCEND, Lille, France
| | - Giovanni B. Frisoni
- Lab of Alzheimer's Neuroimaging and Epidemiology, IRCCS Istituto Centro San Giovanni di Dio Fatebenefratelli, Brescia, Italy
| | - Catherine J. Mummery
- Dementia Research Centre, Institute of Neurology, University College London, London, United Kingdom
| | - Rik Vandenberghe
- Neurology Service University Hospitals Leuven, Leuven, Belgium
- Laboratory for Cognitive Neurology, Department of Neurosciences, Leuven Brain Institute, Leuven, Belgium
| | - Isabelle Le Ber
- Paris Brain Institute, Sorbonne University, Paris, France
- Neurology Department, Reference Centre for Rare or Early Dementias, Paris, France
| | - Didier Hannequin
- CHU Rouen Normandie, Service de Neurologie, Rouen University Hospital, Rouen, France
| | - Scott M. McGinnis
- Department of Neurology, Massachusetts General Hospital, Charlestown
| | - Sophie Auriacombe
- Neurology Department, CHU Bordeaux, Bordeaux Hospital, Bordeaux, France
| | - Marco Onofrj
- Department of Neuroscience Imaging, University G d'Annunzio of Chieti-Pescara, Chieti, Italy
| | | | | | | | - Jacob Hesterman
- Institute for Neurodegenerative Disorders, New Haven, Connecticut
| | - Ken Marek
- Invicro, A Konica Minolta Company, Boston, Massachusetts
- Institute for Neurodegenerative Disorders, New Haven, Connecticut
| | - Beth Ann Haynes
- FORUM Pharmaceuticals Incorporated, Waltham, Massachusetts
- Alector, South San Francisco, California
| | | | - Gerhard Koenig
- FORUM Pharmaceuticals Incorporated, Waltham, Massachusetts
- Arkuda Therapeutics, Watertown, Massachusetts
| | - Dana Hilt
- Arkuda Therapeutics, Watertown, Massachusetts
- Lysosomal Therapeutics, Cambridge, Massachusetts
- Frequency Therapeutics, Farmington, Connecticut
| | - Hans Moebius
- FORUM Pharmaceuticals Incorporated, Waltham, Massachusetts
- Athira Pharma Inc, Seattle, Washington
| | - Adam L. Boxer
- Memory and Aging Center, Department of Neurology, University of California at San Francisco, Weill Institute for Neurosciences, San Francisco
| |
Collapse
|
17
|
Terryn J, Verfaillie CM, Van Damme P. Tweaking Progranulin Expression: Therapeutic Avenues and Opportunities. Front Mol Neurosci 2021; 14:713031. [PMID: 34366786 PMCID: PMC8343103 DOI: 10.3389/fnmol.2021.713031] [Citation(s) in RCA: 23] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/07/2021] [Indexed: 01/01/2023] Open
Abstract
Frontotemporal dementia (FTD) is a neurodegenerative disease, leading to behavioral changes and language difficulties. Heterozygous loss-of-function mutations in progranulin (GRN) induce haploinsufficiency of the protein and are associated with up to one-third of all genetic FTD cases worldwide. While the loss of GRN is primarily associated with neurodegeneration, the biological functions of the secreted growth factor-like protein are more diverse, ranging from wound healing, inflammation, vasculogenesis, and metabolic regulation to tumor cell growth and metastasis. To date, no disease-modifying treatments exist for FTD, but different therapeutic approaches to boost GRN levels in the central nervous system are currently being developed (including AAV-mediated GRN gene delivery as well as anti-SORT1 antibody therapy). In this review, we provide an overview of the multifaceted regulation of GRN levels and the corresponding therapeutic avenues. We discuss the opportunities, advantages, and potential drawbacks of the diverse approaches. Additionally, we highlight the therapeutic potential of elevating GRN levels beyond patients with loss-of-function mutations in GRN.
Collapse
Affiliation(s)
- Joke Terryn
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium.,Laboratory of Neurobiology, Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Catherine M Verfaillie
- Department of Development and Regeneration, Interdepartmental Stem Cell Institute, KU Leuven-University of Leuven, Leuven, Belgium
| | - Philip Van Damme
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium.,Laboratory of Neurobiology, Center for Brain and Disease Research, VIB, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
18
|
Lanter JC, Chen AYP, Williamson T, Koenig G, Blain JF, Burnett DA. Discovery of quinuclidine modulators of cellular progranulin. Bioorg Med Chem Lett 2021; 47:128209. [PMID: 34153473 DOI: 10.1016/j.bmcl.2021.128209] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 11/16/2022]
Abstract
Phenotypic screening of an annotated small molecule library identified the quinuclidine tetrahydroisoquinoline solifenacin (1) as a robust enhancer of progranulin secretion with single digit micromolar potency in a murine microglial (BV-2) cell line. Subsequent SAR development led to the identification of 29 with a 38-fold decrease in muscarinic receptor antagonist activity and a 10-fold improvement in BV-2 potency.
Collapse
Affiliation(s)
- James C Lanter
- Arkuda Therapeutics, 200 Arsenal Yards Blvd Suite 220, Watertown, MA 02472, USA
| | - Angela Y-P Chen
- Arkuda Therapeutics, 200 Arsenal Yards Blvd Suite 220, Watertown, MA 02472, USA
| | - Toni Williamson
- Arkuda Therapeutics, 200 Arsenal Yards Blvd Suite 220, Watertown, MA 02472, USA
| | - Gerhard Koenig
- Arkuda Therapeutics, 200 Arsenal Yards Blvd Suite 220, Watertown, MA 02472, USA
| | - Jean-François Blain
- Arkuda Therapeutics, 200 Arsenal Yards Blvd Suite 220, Watertown, MA 02472, USA
| | - Duane A Burnett
- Arkuda Therapeutics, 200 Arsenal Yards Blvd Suite 220, Watertown, MA 02472, USA
| |
Collapse
|
19
|
Vieira SRL, Morris HR. Neurodegenerative Disease Risk in Carriers of Autosomal Recessive Disease. Front Neurol 2021; 12:679927. [PMID: 34149605 PMCID: PMC8211888 DOI: 10.3389/fneur.2021.679927] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/14/2021] [Indexed: 01/19/2023] Open
Abstract
Genetics has driven significant discoveries in the field of neurodegenerative diseases (NDDs). An emerging theme in neurodegeneration warrants an urgent and comprehensive update: that carrier status of early-onset autosomal recessive (AR) disease, typically considered benign, is associated with an increased risk of a spectrum of late-onset NDDs. Glucosylceramidase beta (GBA1) gene mutations, responsible for the AR lysosomal storage disorder Gaucher disease, are a prominent example of this principle, having been identified as an important genetic risk factor for Parkinson disease. Genetic analyses have revealed further examples, notably GRN, TREM2, EIF2AK3, and several other LSD and mitochondria function genes. In this Review, we discuss the evidence supporting the strikingly distinct allele-dependent clinical phenotypes observed in carriers of such gene mutations and its impact on the wider field of neurodegeneration.
Collapse
Affiliation(s)
| | - Huw R. Morris
- Department of Clinical and Movement Neurosciences, University College London, Queen Square Institute of Neurology, London, United Kingdom
| |
Collapse
|
20
|
Wang XM, Zeng P, Fang YY, Zhang T, Tian Q. Progranulin in neurodegenerative dementia. J Neurochem 2021; 158:119-137. [PMID: 33930186 DOI: 10.1111/jnc.15378] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2020] [Revised: 03/28/2021] [Accepted: 04/16/2021] [Indexed: 01/21/2023]
Abstract
Long-term or severe lack of protective factors is important in the pathogenesis of neurodegenerative dementia. Progranulin (PGRN), a neurotrophic factor expressed mainly in neurons and microglia, has various neuroprotective effects such as anti-inflammatory effects, promoting neuron survival and neurite growth, and participating in normal lysosomal function. Mutations in the PGRN gene (GRN) have been found in several neurodegenerative dementias, including frontotemporal lobar degeneration (FTLD) and Alzheimer's disease (AD). Herein, PGRN deficiency and PGRN hydrolytic products (GRNs) in the pathological changes related to dementia, including aggregation of tau and TAR DNA-binding protein 43 (TDP-43), amyloid-β (Aβ) overproduction, neuroinflammation, lysosomal dysfunction, neuronal death, and synaptic deficit have been summarized. Furthermore, as some therapeutic strategies targeting PGRN have been developed in various models, we highlighted PGRN as a potential anti-neurodegeneration target in dementia.
Collapse
Affiliation(s)
- Xiao-Ming Wang
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry, Huazhong University of Science and Technology, Wuhan, China
| | - Peng Zeng
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry, Huazhong University of Science and Technology, Wuhan, China
| | - Ying-Yan Fang
- Hubei Key Laboratory for Kidney Disease Pathogenesis and Intervention, Hubei Polytechnic University School of Medicine, Huangshi, China
| | - Teng Zhang
- Department of Neurology, Shanxian Central Hospital, The Affiliated Huxi Hospital of Jining Medical College, Heze, China
| | - Qing Tian
- Department of Pathology and Pathophysiology, School of Basic Medicine, Tongji Medical College, Key Laboratory of Neurological Disease of National Education Ministry, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
21
|
Hummel R, Lang M, Walderbach S, Wang Y, Tegeder I, Gölz C, Schäfer MKE. Single intracerebroventricular progranulin injection adversely affects the blood-brain barrier in experimental traumatic brain injury. J Neurochem 2021; 158:342-357. [PMID: 33899947 DOI: 10.1111/jnc.15375] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 04/17/2021] [Accepted: 04/18/2021] [Indexed: 12/23/2022]
Abstract
Progranulin (PGRN) is a neurotrophic and anti-inflammatory factor with protective effects in animal models of ischemic stroke, subarachnoid hemorrhage, and traumatic brain injury (TBI). Administration of recombinant (r) PGRN prevents exaggerated brain pathology after TBI in Grn-deficient mice, suggesting that local injection of recombinant progranulin (rPGRN) provides therapeutic benefit in the acute phase of TBI. To test this hypothesis, we subjected adult male C57Bl/6N mice to the controlled cortical impact model of TBI, administered a single dose of rPGRN intracerebroventricularly (ICV) shortly before the injury, and examined behavioral and biological effects up to 5 days post injury (dpi). The anti-inflammatory bioactivity of rPGRN was confirmed by its capability to inhibit the inflammation-induced hypertrophy of murine primary microglia and astrocytes in vitro. In C57Bl/6N mice, however, ICV administration of rPGRN failed to attenuate behavioral deficits over the 5-day observation period. (Immuno)histological gene and protein expression analyses at 5 dpi did not reveal a therapeutic benefit in terms of brain injury size, brain inflammation, glia activation, cell numbers in neurogenic niches, and neuronal damage. Instead, we observed a failure of TBI-induced mRNA upregulation of the tight junction protein occludin and increased extravasation of serum immunoglobulin G into the brain parenchyma at 5 dpi. In conclusion, single ICV administration of rPGRN had not the expected protective effects in the acute phase of murine TBI, but appeared to cause an aggravation of blood-brain barrier disruption. The data raise questions about putative PGRN-boosting approaches in other types of brain injuries and disease.
Collapse
Affiliation(s)
- Regina Hummel
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Manuel Lang
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Simona Walderbach
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Yong Wang
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University Frankfurt, Frankfurt, Germany
| | - Christina Gölz
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Focus Program Translational Neurosciences (FTN) of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Research Center for Immunotherapy (FZI) of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
22
|
Athira KV, Sadanandan P, Chakravarty S. Repurposing Vorinostat for the Treatment of Disorders Affecting Brain. Neuromolecular Med 2021; 23:449-465. [PMID: 33948878 DOI: 10.1007/s12017-021-08660-4] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2020] [Accepted: 04/09/2021] [Indexed: 12/19/2022]
Abstract
Based on the findings in recent years, we summarize the therapeutic potential of vorinostat (VOR), the first approved histone deacetylase (HDAC) inhibitor, in disorders of brain, and strategies to improve drug efficacy and reduce side effects. Scientific evidences provide a strong case for the therapeutic utility of VOR in various disorders affecting brain, including stroke, Alzheimer's disease, frontotemporal dementia, Parkinson's disease, Huntington's disease, amyotrophic lateral sclerosis, spinal muscular atrophy, X-linked adrenoleukodystrophy, epilepsy, Niemann-Pick type C disease, and neuropsychiatric disorders. Further elucidation of the neuroprotective and neurorestorative properties of VOR using proper clinical study designs could provide momentum towards its clinical application. To improve the therapeutic prospect, concerns on systemic toxicity and off-target actions need to be addressed along with the improvement in formulation and delivery aspects, especially with respect to solubility, permeability, and pharmacokinetic properties. Newer approaches in this regard include poly(ethylene glycol)-b-poly(DL-lactic acid) micelles, VOR-pluronic F127 micelles, encapsulation of iron complexes of VOR into PEGylated liposomes, human serum albumin bound VOR nanomedicine, magnetically guided layer-by-layer assembled nanocarriers, as well as convection-enhanced delivery. Even though targeting specific class or isoform of HDAC is projected as advantageous over pan-HDAC inhibitor like VOR, in terms of adverse effects and efficacy, till clinical validation, the idea is debated. As the VOR treatment-related adverse changes are mostly found reversible, further optimization of the therapeutic strategies with respect to dose, dosage regimen, and formulations of VOR could propel its clinical prospects.
Collapse
Affiliation(s)
- K V Athira
- Department of Pharmacology, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Amrita Institute of Medical Sciences Health Sciences Campus, Kochi, 682 041, Kerala, India.
| | - Prashant Sadanandan
- Department of Pharmaceutical Chemistry & Analysis, Amrita School of Pharmacy, Amrita Vishwa Vidyapeetham, Amrita Institute of Medical Sciences Health Sciences Campus, Kochi, 682 041, Kerala, India
| | - Sumana Chakravarty
- Applied Biology Division, CSIR- Indian Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad, 500007, Telangana, India.
| |
Collapse
|
23
|
Khoury R, Liu Y, Sheheryar Q, Grossberg GT. Pharmacotherapy for Frontotemporal Dementia. CNS Drugs 2021; 35:425-438. [PMID: 33840052 DOI: 10.1007/s40263-021-00813-0] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Accepted: 03/26/2021] [Indexed: 12/11/2022]
Abstract
Frontotemporal dementia is a heterogeneous spectrum of neurodegenerative disorders. The neuropathological inclusions are tau proteins, TAR DNA binding protein 43 kDa-TDP-43, or fused in sarcoma-ubiquitinated inclusions. Genetically, several autosomal mutations account for the heritability of the disorder. Phenotypically, frontotemporal dementia can present with a behavioral variant or a language variant called primary progressive aphasia. To date, there are no approved symptomatic or disease-modifying treatments for frontotemporal dementia. Currently used therapies are supported by low-level of evidence (mostly uncontrolled) studies. The off-label use of drugs is also limited by their side-effect profile including an increased risk of confusion, parkinsonian symptoms, and risk of mortality. Emerging disease-modifying treatments currently target the progranulin and the expansion on chromosome 9 open reading frame 72 genes as well as tau deposits. Advancing our understanding of the pathophysiology of the disease and improving the design of future clinical trials are much needed to optimize the chances to obtain positive outcomes.
Collapse
Affiliation(s)
- Rita Khoury
- Department of Psychiatry and Clinical Psychology, Saint Georges Hospital University Medical Center, Youssef Sursock Street, PO Box 166378, Beirut, Lebanon. .,Faculty of Medicine, University of Balamand, Beirut, Lebanon. .,Department of Psychiatry and Behavioral Neuroscience, Saint Louis University School of Medicine, Saint Louis, MO, USA.
| | - Yu Liu
- Department of Psychiatry and Behavioral Neuroscience, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - Quratulanne Sheheryar
- Department of Psychiatry and Behavioral Neuroscience, Saint Louis University School of Medicine, Saint Louis, MO, USA
| | - George T Grossberg
- Department of Psychiatry and Behavioral Neuroscience, Saint Louis University School of Medicine, Saint Louis, MO, USA
| |
Collapse
|
24
|
Giunta M, Solje E, Gardoni F, Borroni B, Benussi A. Experimental Disease-Modifying Agents for Frontotemporal Lobar Degeneration. J Exp Pharmacol 2021; 13:359-376. [PMID: 33790662 PMCID: PMC8005747 DOI: 10.2147/jep.s262352] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/22/2021] [Accepted: 03/09/2021] [Indexed: 12/11/2022] Open
Abstract
Frontotemporal dementia is a clinically, genetically and pathologically heterogeneous neurodegenerative disorder, enclosing a wide range of different pathological entities, associated with the accumulation of proteins such as tau and TPD-43. Characterized by a high hereditability, mutations in three main genes, MAPT, GRN and C9orf72, can drive the neurodegenerative process. The connection between different genes and proteinopathies through specific mechanisms has shed light on the pathophysiology of the disease, leading to the identification of potential pharmacological targets. New experimental strategies are emerging, in both preclinical and clinical settings, which focus on small molecules rather than gene therapy. In this review, we provide an insight into the aberrant mechanisms leading to FTLD-related proteinopathies and discuss recent therapies with the potential to ameliorate neurodegeneration and disease progression.
Collapse
Affiliation(s)
- Marcello Giunta
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Eino Solje
- Institute of Clinical Medicine - Neurology, University of Eastern Finland, Kuopio, Finland
| | - Fabrizio Gardoni
- Department of Pharmacological and Biomolecular Sciences, University of Milan, Milan, Italy
| | - Barbara Borroni
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Alberto Benussi
- Neurology Unit, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
25
|
Mossink B, Negwer M, Schubert D, Nadif Kasri N. The emerging role of chromatin remodelers in neurodevelopmental disorders: a developmental perspective. Cell Mol Life Sci 2021; 78:2517-2563. [PMID: 33263776 PMCID: PMC8004494 DOI: 10.1007/s00018-020-03714-5] [Citation(s) in RCA: 57] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2020] [Revised: 11/04/2020] [Accepted: 11/16/2020] [Indexed: 12/13/2022]
Abstract
Neurodevelopmental disorders (NDDs), including intellectual disability (ID) and autism spectrum disorders (ASD), are a large group of disorders in which early insults during brain development result in a wide and heterogeneous spectrum of clinical diagnoses. Mutations in genes coding for chromatin remodelers are overrepresented in NDD cohorts, pointing towards epigenetics as a convergent pathogenic pathway between these disorders. In this review we detail the role of NDD-associated chromatin remodelers during the developmental continuum of progenitor expansion, differentiation, cell-type specification, migration and maturation. We discuss how defects in chromatin remodelling during these early developmental time points compound over time and result in impaired brain circuit establishment. In particular, we focus on their role in the three largest cell populations: glutamatergic neurons, GABAergic neurons, and glia cells. An in-depth understanding of the spatiotemporal role of chromatin remodelers during neurodevelopment can contribute to the identification of molecular targets for treatment strategies.
Collapse
Affiliation(s)
- Britt Mossink
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, Geert Grooteplein 10, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, 6500 HB, Nijmegen, The Netherlands
| | - Moritz Negwer
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, Geert Grooteplein 10, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, 6500 HB, Nijmegen, The Netherlands
| | - Dirk Schubert
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, 6500 HB, Nijmegen, The Netherlands
| | - Nael Nadif Kasri
- Department of Human Genetics, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, Geert Grooteplein 10, P.O. Box 9101, 6500 HB, Nijmegen, The Netherlands.
- Department of Cognitive Neuroscience, Radboudumc, Donders Institute for Brain, Cognition and Behaviour, 6500 HB, Nijmegen, The Netherlands.
| |
Collapse
|
26
|
Shetty R, Kumar NR, Subramani M, Krishna L, Murugeswari P, Matalia H, Khamar P, Dadachanji ZV, Mohan RR, Ghosh A, Das D. Safety and efficacy of combination of suberoylamilide hydroxyamic acid and mitomycin C in reducing pro-fibrotic changes in human corneal epithelial cells. Sci Rep 2021; 11:4392. [PMID: 33623133 PMCID: PMC7902619 DOI: 10.1038/s41598-021-83881-y] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2020] [Accepted: 02/01/2021] [Indexed: 12/14/2022] Open
Abstract
Corneal haze post refractive surgery is prevented by mitomycin c (MMC) treatment though it can lead to corneal endothelial damage, persistent epithelial defects and necrosis of cells. Suberanilohydroxamic acid (SAHA) however has been proposed to prevent corneal haze without any adverse effects. For clinical application we have investigated the short and long term outcome of cells exposed to SAHA. Human donor cornea, cultured limbal epithelial cells, corneal rims and lenticules were incubated with SAHA and MMC. The cells/tissue was then analyzed by RT-qPCR, immunofluorescence and western blot for markers of apoptosis and fibrosis. The results reveal that short term exposure of SAHA and SAHA + MMC reduced apoptosis levels and increased αSMA expression compared to those treated with MMC. Epithelial cells derived from cultured corneal rim that were incubated with the MMC, SAHA or MMC + SAHA revealed enhanced apoptosis, reduced levels of CK3/CK12, ∆NP63 and COL4A compared to other treatments. In SAHA treated lenticules TGFβ induced fibrosis was reduced. The results imply that MMC treatment for corneal haze has both short term and long term adverse effects on cells and the cellular properties. However, a combinatorial treatment of SAHA + MMC prevents expression of corneal fibrotic markers without causing any adverse effect on cellular properties.
Collapse
Affiliation(s)
- Rohit Shetty
- Department of Cornea and Refractive Surgery, Narayana Nethralaya Eye Hospital, Bangalore, Karnataka, India
| | - Nimisha Rajiv Kumar
- GROW Laboratory, Narayana Nethralaya Post Graduate Institute of Ophthalmology, Narayana Nethralaya Foundation, Narayana Nethralaya, Narayana Health City, Bommasandra, , Bangalore, Karnataka, 560 099, India
| | - Murali Subramani
- Stem Cell Research Lab, GROW Laboratory, Narayana Nethralaya Foundation, Narayana Nethralaya, Bangalore, Karnataka, India
| | - Lekshmi Krishna
- Stem Cell Research Lab, GROW Laboratory, Narayana Nethralaya Foundation, Narayana Nethralaya, Bangalore, Karnataka, India
| | - Ponnalagu Murugeswari
- Stem Cell Research Lab, GROW Laboratory, Narayana Nethralaya Foundation, Narayana Nethralaya, Bangalore, Karnataka, India
| | - Himanshu Matalia
- Department of Cornea and Refractive Surgery, Narayana Nethralaya Eye Hospital, Bangalore, Karnataka, India
| | - Pooja Khamar
- Department of Cornea and Refractive Surgery, Narayana Nethralaya Eye Hospital, Bangalore, Karnataka, India
| | - Zelda V Dadachanji
- Department of Cornea and Refractive Surgery, Narayana Nethralaya Eye Hospital, Bangalore, Karnataka, India
| | - Rajiv R Mohan
- Department of Veterinary Medicine and Surgery, University of Missouri, Columbia, MO, 65211, USA. .,Mason Eye Institute, School of Medicine, University of Missouri, Columbia, MO, 65212, USA. .,Harry S Truman Veterans' Memorial Hospital, Columbia, MO, 65201, USA.
| | - Arkasubhra Ghosh
- GROW Laboratory, Narayana Nethralaya Post Graduate Institute of Ophthalmology, Narayana Nethralaya Foundation, Narayana Nethralaya, Narayana Health City, Bommasandra, , Bangalore, Karnataka, 560 099, India.
| | - Debashish Das
- Stem Cell Research Lab, GROW Laboratory, Narayana Nethralaya Foundation, Narayana Nethralaya, Bangalore, Karnataka, India.
| |
Collapse
|
27
|
FTLD Treatment: Current Practice and Future Possibilities. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1281:297-310. [PMID: 33433882 DOI: 10.1007/978-3-030-51140-1_18] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/21/2023]
Abstract
While behavioral variant frontotemporal dementia (bvFTD) and primary progressive aphasia (PPA) remain unrelenting and universally fatal conditions, there is a framework for supportive treatment in patients diagnosed with these frontotemporal dementia (FTD) syndromes and the larger spectrum of clinical syndromes associated with frontotemporal lobar degeneration (FTLD) pathology on autopsy. A managing physician has an important role in weighing therapeutic options, organizing caregiver support, and framing long-term expectations for patients and caregivers. Additionally, a dedicated neurologist may assist patients and caregivers in navigating a growing range of FTD research, including exciting opportunities in clinical therapeutic trials. This chapter will review current therapeutic options for patients with bvFTD and PPA and detail the landscape of potential new disease-modifying therapies targeting the pathophysiology or FTLD.
Collapse
|
28
|
Miyakawa S, Sakuma H, Warude D, Asanuma S, Arimura N, Yoshihara T, Tavares D, Hata A, Ida K, Hori Y, Okuzono Y, Yamamoto S, Iida K, Shimizu H, Kondo S, Sato S. Anti-sortilin1 Antibody Up-Regulates Progranulin via Sortilin1 Down-Regulation. Front Neurosci 2020; 14:586107. [PMID: 33384578 PMCID: PMC7770147 DOI: 10.3389/fnins.2020.586107] [Citation(s) in RCA: 17] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2020] [Accepted: 11/16/2020] [Indexed: 12/15/2022] Open
Abstract
Progranulin (PGRN) haploinsufficiency associated with loss-of-function mutations in the granulin gene causes frontotemporal dementia (FTD). This suggests that increasing PGRN levels could have promising therapeutic implications for patients carrying GRN mutations. In this study, we explored the therapeutic potential of sortilin1 (SORT1), a clearance receptor of PGRN, by generating and characterizing monoclonal antibodies against SORT1. Anti-SORT1 monoclonal antibodies were generated by immunizing Sort1 knockout mice with SORT1 protein. The antibodies were classified into 7 epitope bins based on their competitive binding to the SORT1 protein and further defined by epitope bin-dependent characteristics, including SORT1-PGRN blocking, SORT1 down-regulation, and binding to human and mouse SORT1. We identified a positive correlation between PGRN up-regulation and SORT1 down-regulation. Furthermore, we also characterized K1-67 antibody via SORT1 down-regulation and binding to mouse SORT1 in vivo and confirmed that K1-67 significantly up-regulated PGRN levels in plasma and brain interstitial fluid of mice. These data indicate that SORT1 down-regulation is a key mechanism in increasing PGRN levels via anti-SORT1 antibodies and suggest that SORT1 is a potential target to correct PGRN reduction, such as that in patients with FTD caused by GRN mutation.
Collapse
Affiliation(s)
- Shuuichi Miyakawa
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hiroyuki Sakuma
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Dnyaneshwar Warude
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Satomi Asanuma
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Naoto Arimura
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Tomoki Yoshihara
- Global Biologics Research, Takeda Pharmaceutical Company Limited, Cambridge, MA, United States
| | - Daniel Tavares
- Global Biologics Research, Takeda Pharmaceutical Company Limited, Cambridge, MA, United States
| | - Akito Hata
- Global Biologics Research, Takeda Pharmaceutical Company Limited, Cambridge, MA, United States
| | - Koh Ida
- Global Biologics Research, Takeda Pharmaceutical Company Limited, Cambridge, MA, United States
| | - Yuri Hori
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Yuumi Okuzono
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Syunsuke Yamamoto
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Koichi Iida
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Hisao Shimizu
- Drug Metabolism and Pharmacokinetics Research Laboratories, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Shinichi Kondo
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| | - Shuji Sato
- Immunology Unit, Research, Takeda Pharmaceutical Company Limited, Fujisawa, Japan
| |
Collapse
|
29
|
Zhu GJ, Gong S, Ma DB, Tao T, He WQ, Zhang L, Wang F, Qian XY, Zhou H, Fan C, Wang P, Chen X, Zhao W, Sun J, Chen H, Wang Y, Gao X, Zuo J, Zhu MS, Gao X, Wan G. Aldh inhibitor restores auditory function in a mouse model of human deafness. PLoS Genet 2020; 16:e1009040. [PMID: 32970669 PMCID: PMC7553308 DOI: 10.1371/journal.pgen.1009040] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2020] [Revised: 10/13/2020] [Accepted: 08/10/2020] [Indexed: 02/06/2023] Open
Abstract
Genetic hearing loss is a common health problem with no effective therapy currently available. DFNA15, caused by mutations of the transcription factor POU4F3, is one of the most common forms of autosomal dominant non-syndromic deafness. In this study, we established a novel mouse model of the human DFNA15 deafness, with a Pou4f3 gene mutation (Pou4f3Δ) identical to that found in a familial case of DFNA15. The Pou4f3(Δ/+) mice suffered progressive deafness in a similar manner to the DFNA15 patients. Hair cells in the Pou4f3(Δ/+) cochlea displayed significant stereociliary and mitochondrial pathologies, with apparent loss of outer hair cells. Progression of hearing and outer hair cell loss of the Pou4f3(Δ/+) mice was significantly modified by other genetic and environmental factors. Using Pou4f3(-/+) heterozygous knockout mice, we also showed that DFNA15 is likely caused by haploinsufficiency of the Pou4f3 gene. Importantly, inhibition of retinoic acid signaling by the aldehyde dehydrogenase (Aldh) and retinoic acid receptor inhibitors promoted Pou4f3 expression in the cochlear tissue and suppressed the progression of hearing loss in the mutant mice. These data demonstrate Pou4f3 haploinsufficiency as the main underlying cause of human DFNA15 deafness and highlight the therapeutic potential of Aldh inhibitors for treatment of progressive hearing loss. More than 50% of deafness cases are due to genetic defects with no treatment available. DFNA15, caused by mutations of the transcription factor POU4F3, is one of the most common types of autosomal dominant non-syndromic deafness. Here, we established a novel mouse model with the exact Pou4f3 mutation identified in human patients. The mutant mouse display similar auditory pathophysiology as human patients and exhibit multiple hair cell abnormalities. The onset and severity of hearing loss in the mouse model is highly modifiable to environmental factors, such as aging, noise exposure or genetic backgrounds. Using a new knockout mouse model, we found Pou4f3 haploinsufficiency as the underlying mechanism of human DFNA15. Importantly, we identified Aldh inhibitor as a potent small molecule for upregulation of Pou4f3 and treatment of hearing loss in the mutant mouse. The identification of Aldh inhibitor for treatment of DFNA15 deafness represents a major advance in the unmet medical need for this common form of progressive hearing loss.
Collapse
Affiliation(s)
- Guang-Jie Zhu
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
| | - Sihao Gong
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
| | - Deng-Bin Ma
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
| | - Tao Tao
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
| | - Wei-Qi He
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and Cambridge-Suda (CAM-SU) Genomic Resource Center, Medical College of Soochow University, Suzhou, China
| | - Linqing Zhang
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
| | - Fang Wang
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
| | - Xiao-Yun Qian
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
| | - Han Zhou
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
| | - Chi Fan
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
| | - Pei Wang
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
| | - Xin Chen
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
| | - Wei Zhao
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
| | - Jie Sun
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
| | - Huaqun Chen
- College of Life Science, Nanjing Normal University, Nanjing, China
| | - Ye Wang
- Nanjing MuCyte Biotechnology Co., Ltd., Nanjing, China
| | - Xiang Gao
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
| | - Jian Zuo
- Department of Biomedical Sciences, School of Medicine, Creighton University, United States of America
| | - Min-Sheng Zhu
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
- * E-mail: (MSZ); (XG); (GW)
| | - Xia Gao
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
- * E-mail: (MSZ); (XG); (GW)
| | - Guoqiang Wan
- Department of Otorhinolaryngology, Provincial Key Discipline of the affiliated Drum Tower Hospital of Nanjing University and Model Animal Research Center, MOE Key Laboratory of Model Animal for Disease Studies, School of Medicine, Nanjing University, Nanjing, China
- Institute for Brain Sciences, Nanjing University, Nanjing, China
- * E-mail: (MSZ); (XG); (GW)
| |
Collapse
|
30
|
Telpoukhovskaia MA, Liu K, Sayed FA, Etchegaray JI, Xie M, Zhan L, Li Y, Zhou Y, Le D, Bahr BA, Bogyo M, Ding S, Gan L. Discovery of small molecules that normalize the transcriptome and enhance cysteine cathepsin activity in progranulin-deficient microglia. Sci Rep 2020; 10:13688. [PMID: 32792571 PMCID: PMC7426857 DOI: 10.1038/s41598-020-70534-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 07/17/2020] [Indexed: 12/11/2022] Open
Abstract
Patients with frontotemporal dementia (FTD) resulting from granulin (GRN) haploinsufficiency have reduced levels of progranulin and exhibit dysregulation in inflammatory and lysosomal networks. Microglia produce high levels of progranulin, and reduction of progranulin in microglia alone is sufficient to recapitulate inflammation, lysosomal dysfunction, and hyperproliferation in a cell-autonomous manner. Therefore, targeting microglial dysfunction caused by progranulin insufficiency represents a potential therapeutic strategy to manage neurodegeneration in FTD. Limitations of current progranulin-enhancing strategies necessitate the discovery of new targets. To identify compounds that can reverse microglial defects in Grn-deficient mouse microglia, we performed a compound screen coupled with high throughput sequencing to assess key transcriptional changes in inflammatory and lysosomal pathways. Positive hits from this initial screen were then further narrowed down based on their ability to rescue cathepsin activity, a critical biochemical readout of lysosomal capacity. The screen identified nor-binaltorphimine dihydrochloride (nor-BNI) and dibutyryl-cAMP, sodium salt (DB-cAMP) as two phenotypic modulators of progranulin deficiency. In addition, nor-BNI and DB-cAMP also rescued cell cycle abnormalities in progranulin-deficient cells. These data highlight the potential of a transcription-based platform for drug screening, and advance two novel lead compounds for FTD.
Collapse
Affiliation(s)
- Maria A Telpoukhovskaia
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA.,Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Kai Liu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, 94158, USA
| | - Faten A Sayed
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA.,Department of Neurology, University of California, San Francisco, CA, 94158, USA.,Neuroscience Graduate Program, University of California, San Francisco, CA, 94158, USA
| | | | - Min Xie
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA.,Gladstone Institute of Cardiovascular Disease, San Francisco, CA, 94158, USA
| | - Lihong Zhan
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA.,Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Yaqiao Li
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
| | - Yungui Zhou
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
| | - David Le
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
| | - Ben A Bahr
- Biotechnology Research and Training Center, University of North Carolina At Pembroke, Pembroke, NC, 28372, USA
| | - Matthew Bogyo
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Sheng Ding
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, 94158, USA
| | - Li Gan
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA. .,Department of Neurology, University of California, San Francisco, CA, 94158, USA. .,Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
31
|
Poeta L, Padula A, Attianese B, Valentino M, Verrillo L, Filosa S, Shoubridge C, Barra A, Schwartz CE, Christensen J, van Bokhoven H, Helin K, Lioi MB, Collombat P, Gecz J, Altucci L, Di Schiavi E, Miano MG. Histone demethylase KDM5C is a SAHA-sensitive central hub at the crossroads of transcriptional axes involved in multiple neurodevelopmental disorders. Hum Mol Genet 2020; 28:4089-4102. [PMID: 31691806 DOI: 10.1093/hmg/ddz254] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/19/2019] [Revised: 10/17/2019] [Accepted: 10/21/2019] [Indexed: 12/26/2022] Open
Abstract
A disproportional large number of neurodevelopmental disorders (NDDs) is caused by variants in genes encoding transcription factors and chromatin modifiers. However, the functional interactions between the corresponding proteins are only partly known. Here, we show that KDM5C, encoding a H3K4 demethylase, is at the intersection of transcriptional axes under the control of three regulatory proteins ARX, ZNF711 and PHF8. Interestingly, mutations in all four genes (KDM5C, ARX, ZNF711 and PHF8) are associated with X-linked NDDs comprising intellectual disability as a core feature. in vitro analysis of the KDM5C promoter revealed that ARX and ZNF711 function as antagonist transcription factors that activate KDM5C expression and compete for the recruitment of PHF8. Functional analysis of mutations in these genes showed a correlation between phenotype severity and the reduction in KDM5C transcriptional activity. The KDM5C decrease was associated with a lack of repression of downstream target genes Scn2a, Syn1 and Bdnf in the embryonic brain of Arx-null mice. Aiming to correct the faulty expression of KDM5C, we studied the effect of the FDA-approved histone deacetylase inhibitor suberanilohydroxamic acid (SAHA). In Arx-KO murine ES-derived neurons, SAHA was able to rescue KDM5C depletion, recover H3K4me3 signalling and improve neuronal differentiation. Indeed, in ARX/alr-1-deficient Caenorhabditis elegans animals, SAHA was shown to counteract the defective KDM5C/rbr-2-H3K4me3 signalling, recover abnormal behavioural phenotype and ameliorate neuronal maturation. Overall, our studies indicate that KDM5C is a conserved and druggable effector molecule across a number of NDDs for whom the use of SAHA may be considered a potential therapeutic strategy.
Collapse
Affiliation(s)
- Loredana Poeta
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", National Research Council (CNR), Naples, Italy
| | - Agnese Padula
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", National Research Council (CNR), Naples, Italy.,University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Benedetta Attianese
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", National Research Council (CNR), Naples, Italy
| | - Mariaelena Valentino
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", National Research Council (CNR), Naples, Italy
| | - Lucia Verrillo
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", National Research Council (CNR), Naples, Italy.,University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Stefania Filosa
- Institute of Biosciences and BioResources, National Research Council (CNR), Naples, Italy.,Istituto Neurologico Mediterraneo (Neuromed), Pozzilli, Isernia, Italy
| | - Cheryl Shoubridge
- Intellectual Disability Research, Adelaide Medical School, The University of Adelaide, Adelaide, South Australia, Australia.,Robinson Research Institute, Department of Paediatrics, University of Adelaide, Adelaide, South Australia, Australia
| | - Adriano Barra
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", National Research Council (CNR), Naples, Italy
| | | | - Jesper Christensen
- University of Copenhagen, Biotech Research and Innovation Centre (BRIC), Copenhagen, Denmark.,University of Copenhagen, The Novo Nordisk Foundation Center for Stem Cell Biology (Danstem), Copenhagen, Denmark
| | - Hans van Bokhoven
- Department of Human Genetics, Donders Institute for Brain, Behaviour and Cognition, Radboudumc, Nijmegen, The Netherlands
| | - Kristian Helin
- University of Copenhagen, Biotech Research and Innovation Centre (BRIC), Copenhagen, Denmark.,University of Copenhagen, The Novo Nordisk Foundation Center for Stem Cell Biology (Danstem), Copenhagen, Denmark
| | | | | | - Jozef Gecz
- Faculty of Health and Medical Sciences, The University of Adelaide, Adelaide, Australia
| | - Lucia Altucci
- University of Campania Luigi Vanvitelli, Caserta, Italy
| | - Elia Di Schiavi
- Institute of Biosciences and BioResources, National Research Council (CNR), Naples, Italy
| | - Maria Giuseppina Miano
- Institute of Genetics and Biophysics "Adriano Buzzati-Traverso", National Research Council (CNR), Naples, Italy
| |
Collapse
|
32
|
Development of disease-modifying drugs for frontotemporal dementia spectrum disorders. Nat Rev Neurol 2020; 16:213-228. [PMID: 32203398 DOI: 10.1038/s41582-020-0330-x] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/14/2020] [Indexed: 02/06/2023]
Abstract
Frontotemporal dementia (FTD) encompasses a spectrum of clinical syndromes characterized by progressive executive, behavioural and language dysfunction. The various FTD spectrum disorders are associated with brain accumulation of different proteins: tau, the transactive response DNA binding protein of 43 kDa (TDP43), or fused in sarcoma (FUS) protein, Ewing sarcoma protein and TATA-binding protein-associated factor 15 (TAF15) (collectively known as FET proteins). Approximately 60% of patients with FTD have autosomal dominant mutations in C9orf72, GRN or MAPT genes. Currently available treatments are symptomatic and provide limited benefit. However, the increased understanding of FTD pathogenesis is driving the development of potential disease-modifying therapies. Most of these drugs target pathological tau - this category includes tau phosphorylation inhibitors, tau aggregation inhibitors, active and passive anti-tau immunotherapies, and MAPT-targeted antisense oligonucleotides. Some of these therapeutic approaches are being tested in phase II clinical trials. Pharmacological approaches that target the effects of GRN and C9orf72 mutations are also in development. Key results of large clinical trials will be available in a few years. However, clinical trials in FTD pose several challenges, and the development of specific brain imaging and molecular biomarkers could facilitate the recruitment of clinically homogenous groups to improve the chances of positive clinical trial results.
Collapse
|
33
|
Frew J, Baradaran-Heravi A, Balgi AD, Wu X, Yan TD, Arns S, Shidmoossavee FS, Tan J, Jaquith JB, Jansen-West KR, Lynn FC, Gao FB, Petrucelli L, Feldman HH, Mackenzie IR, Roberge M, Nygaard HB. Premature termination codon readthrough upregulates progranulin expression and improves lysosomal function in preclinical models of GRN deficiency. Mol Neurodegener 2020; 15:21. [PMID: 32178712 PMCID: PMC7075020 DOI: 10.1186/s13024-020-00369-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2020] [Accepted: 02/24/2020] [Indexed: 02/07/2023] Open
Abstract
Background Frontotemporal lobar degeneration (FTLD) is a devastating and progressive disorder, and a common cause of early onset dementia. Progranulin (PGRN) haploinsufficiency due to autosomal dominant mutations in the progranulin gene (GRN) is an important cause of FTLD (FTLD-GRN), and nearly a quarter of these genetic cases are due to a nonsense mutation. Premature termination codons (PTC) can be therapeutically targeted by compounds allowing readthrough, and aminoglycoside antibiotics are known to be potent PTC readthrough drugs. Restoring endogenous PGRN through PTC readthrough has not previously been explored as a therapeutic intervention in FTLD. Methods We studied whether the aminoglycoside G418 could increase PGRN expression in HEK293 and human induced pluripotent stem cell (hiPSC)-derived neurons bearing the heterozygous S116X, R418X, and R493X pathogenic GRN nonsense mutations. We further tested a novel substituted phthalimide PTC readthrough enhancer in combination with G418 in our cellular models. We next generated a homozygous R493X knock-in hiPSC isogenic line (R493X−/− KI), assessing whether combination treatment in hiPSC-derived neurons and astrocytes could increase PGRN and ameliorate lysosomal dysfunction relevant to FTLD-GRN. To provide in vivo proof-of-concept of our approach, we measured brain PGRN after intracerebroventricular administration of G418 in mice expressing the V5-tagged GRN nonsense mutation R493X. Results The R418X and R493X mutant GRN cell lines responded to PTC readthrough with G418, and treatments increased PGRN levels in R493X−/− KI hiPSC-derived neurons and astrocytes. Combining G418 with a PTC readthrough enhancer increased PGRN levels over G418 treatment alone in vitro. PGRN deficiency has been shown to impair lysosomal function, and the mature form of the lysosomal protease cathepsin D is overexpressed in R493X−/− KI neurons. Increasing PGRN through G418-mediated PTC readthrough normalized this abnormal lysosomal phenotype in R493X−/− KI neuronal cultures. A single intracerebroventricular injection of G418 induced GRN PTC readthrough in 6-week-old AAV-GRN-R493X-V5 mice. Conclusions Taken together, our findings suggest that PTC readthrough may be a potential therapeutic strategy for FTLD caused by GRN nonsense mutations.
Collapse
Affiliation(s)
- Jonathan Frew
- Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Alireza Baradaran-Heravi
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Aruna D Balgi
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Xiujuan Wu
- Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Tyler D Yan
- Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Steve Arns
- adMare BioInnovations, Vancouver, British Columbia, Canada
| | | | - Jason Tan
- adMare BioInnovations, Vancouver, British Columbia, Canada
| | | | | | - Francis C Lynn
- Department of Surgery, University of British Columbia, Faculty of Medicine, Vancouver, British Columbia, Canada
| | - Fen-Biao Gao
- Department of Neurology, University of Massachusetts Medical School, Worcester, MA, USA
| | | | - Howard H Feldman
- Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada.,Department of Neurosciences, University of California, San Diego, San Diego, CA, USA
| | - Ian R Mackenzie
- Department of Pathology and Laboratory Medicine, University of British Columbia, Vancouver, British Columbia, Canada
| | - Michel Roberge
- Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, British Columbia, Canada
| | - Haakon B Nygaard
- Division of Neurology, University of British Columbia, Vancouver, British Columbia, Canada.
| |
Collapse
|
34
|
Pharmacological intervention of histone deacetylase enzymes in the neurodegenerative disorders. Life Sci 2020; 243:117278. [PMID: 31926248 DOI: 10.1016/j.lfs.2020.117278] [Citation(s) in RCA: 51] [Impact Index Per Article: 10.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2019] [Revised: 12/31/2019] [Accepted: 01/01/2020] [Indexed: 02/06/2023]
Abstract
Reversal of aging symptoms and related disorders are the challenging task where epigenetic is a crucial player that includes DNA methylation, histone modification; chromatin remodeling and regulation that are linked to the progression of various neurodegenerative disorders (NDDs). Overexpression of various histone deacetylase (HDACs) can activate Glycogen synthase kinase 3 which promotes the hyperphosphorylation of tau and inhibits its degradation. While HDAC is important for maintaining the neuronal morphology and brain homeostasis, at the same time, these enzymes are promoting neurodegeneration, if it is deregulated. Different experimental models have also confirmed the neuroprotective effects caused by HDAC enzymes through the regulation of neuronal apoptosis, inflammatory response, DNA damage, cell cycle regulation, and metabolic dysfunction. Apart from transcriptional regulation, protein-protein interaction, histone post-translational modifications, deacetylation mechanism of non-histone protein and direct association with disease proteins have been linked to neuronal imbalance. Histone deacetylases inhibitors (HDACi) can be able to alter gene expression and shown its efficacy on experimental models, and in clinical trials for NDD's and found to be a very promising therapeutic agent with certain limitation, for instance, non-specific target effect, isoform-selectivity, specificity, and limited number of predicted biomarkers. Herein, we discussed (i) the catalytic mechanism of the deacetylation process of various HDAC's in in vivo and in vitro experimental models, (ii) how HDACs are participating in neuroprotection as well as in neurodegeneration, (iii) a comprehensive role of HDACi in maintaining neuronal homeostasis and (iv) therapeutic role of biomolecules to modulate HDACs.
Collapse
|
35
|
Boxer AL, Gold M, Feldman H, Boeve BF, Dickinson SLJ, Fillit H, Ho C, Paul R, Pearlman R, Sutherland M, Verma A, Arneric SP, Alexander BM, Dickerson BC, Dorsey ER, Grossman M, Huey ED, Irizarry MC, Marks WJ, Masellis M, McFarland F, Niehoff D, Onyike CU, Paganoni S, Panzara MA, Rockwood K, Rohrer JD, Rosen H, Schuck RN, Soares HD, Tatton N. New directions in clinical trials for frontotemporal lobar degeneration: Methods and outcome measures. Alzheimers Dement 2020; 16:131-143. [PMID: 31668596 PMCID: PMC6949386 DOI: 10.1016/j.jalz.2019.06.4956] [Citation(s) in RCA: 42] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Frontotemporal lobar degeneration (FTLD) is the most common form of dementia for those under 60 years of age. Increasing numbers of therapeutics targeting FTLD syndromes are being developed. METHODS In March 2018, the Association for Frontotemporal Degeneration convened the Frontotemporal Degeneration Study Group meeting in Washington, DC, to discuss advances in the clinical science of FTLD. RESULTS Challenges exist for conducting clinical trials in FTLD. Two of the greatest challenges are (1) the heterogeneity of FTLD syndromes leading to difficulties in efficiently measuring treatment effects and (2) the rarity of FTLD disorders leading to recruitment challenges. DISCUSSION New personalized endpoints that are clinically meaningful to individuals and their families should be developed. Personalized approaches to analyzing MRI data, development of new fluid biomarkers and wearable technologies will help to improve the power to detect treatment effects in FTLD clinical trials and enable new, clinical trial designs, possibly leveraged from the experience of oncology trials. A computational visualization and analysis platform that can support novel analyses of combined clinical, genetic, imaging, biomarker data with other novel modalities will be critical to the success of these endeavors.
Collapse
Affiliation(s)
- Adam L. Boxer
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA
| | | | - Howard Feldman
- Department of Neurosciences, University of California San Diego, San Diego, CA
| | | | | | | | - Carole Ho
- Denali Therapeutics, San Francisco, CA
| | | | | | | | | | | | | | | | - Earl Ray Dorsey
- Center for Health and Technology, University of Rochester, Rochester, NY
| | - Murray Grossman
- Department of Neurology, University of Pennsylvania, Philadelphia, PA
| | - Edward D. Huey
- Departments of Psychiatry and Neurology, Columbia University, NY
| | | | - William J. Marks
- Clinical Neurology, Verily Life Sciences, South San Francisco, CA
| | - Mario Masellis
- Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, University of Toronto, ON, Canada; Department of Medicine (Neurology), Sunnybrook Health Sciences Centre, University of Toronto, ON, Canada
| | | | - Debra Niehoff
- Association for Frontotemporal Degeneration, Radnor, PA
| | - Chiadi U. Onyike
- Department Geriatric Psychiatry and Neuropsychiatry, Johns Hopkins University, Baltimore, MD
| | - Sabrina Paganoni
- Healey Center for ALS, Massachusetts General Hospital, Boston, MA
| | | | - Kenneth Rockwood
- Division of Geriatric Medicine, Dalhousie University, Halifax, NS
| | - Jonathan D. Rohrer
- Dementia Research Centre, UCL Institute of Neurology, Queen Square, London, UK
| | - Howard Rosen
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA
| | - Robert N. Schuck
- Office of Clinical Pharmacology, Center for Drug Evaluation and Research, FDA, Silver Spring, MD
| | | | - Nadine Tatton
- Association for Frontotemporal Degeneration, Radnor, PA
| |
Collapse
|
36
|
Fedorova YB. [Current approaches to the treatment of fronto-temporal dementia]. Zh Nevrol Psikhiatr Im S S Korsakova 2019; 119:18-24. [PMID: 31825385 DOI: 10.17116/jnevro201911909218] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Frontotemporal dementias (FTD) are one of the prevalent forms of early neurodegenerative diseases. FTD are characterized by heterogeneous clinical manifestations and syndromes. The current methods of FTD treatment and the clinical trials of new methods of FTD treatment are considered in the article. Biomarkers and their relationships with the results of recently completed clinical trials, as well as future therapeutic perspectives, are reviewed.
Collapse
|
37
|
Mutsaerts HJMM, Mirza SS, Petr J, Thomas DL, Cash DM, Bocchetta M, de Vita E, Metcalfe AWS, Shirzadi Z, Robertson AD, Tartaglia MC, Mitchell SB, Black SE, Freedman M, Tang-Wai D, Keren R, Rogaeva E, van Swieten J, Laforce R, Tagliavini F, Borroni B, Galimberti D, Rowe JB, Graff C, Frisoni GB, Finger E, Sorbi S, de Mendonça A, Rohrer JD, MacIntosh BJ, Masellis M. Cerebral perfusion changes in presymptomatic genetic frontotemporal dementia: a GENFI study. Brain 2019; 142:1108-1120. [PMID: 30847466 PMCID: PMC6439322 DOI: 10.1093/brain/awz039] [Citation(s) in RCA: 38] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2018] [Revised: 12/14/2018] [Accepted: 01/04/2019] [Indexed: 11/12/2022] Open
Abstract
Genetic forms of frontotemporal dementia are most commonly due to mutations in three genes, C9orf72, GRN or MAPT, with presymptomatic carriers from families representing those at risk. While cerebral blood flow shows differences between frontotemporal dementia and other forms of dementia, there is limited evidence of its utility in presymptomatic stages of frontotemporal dementia. This study aimed to delineate the cerebral blood flow signature of presymptomatic, genetic frontotemporal dementia using a voxel-based approach. In the multicentre GENetic Frontotemporal dementia Initiative (GENFI) study, we investigated cross-sectional differences in arterial spin labelling MRI-based cerebral blood flow between presymptomatic C9orf72, GRN or MAPT mutation carriers (n = 107) and non-carriers (n = 113), using general linear mixed-effects models and voxel-based analyses. Cerebral blood flow within regions of interest derived from this model was then explored to identify differences between individual gene carrier groups and to estimate a timeframe for the expression of these differences. The voxel-based analysis revealed a significant inverse association between cerebral blood flow and the expected age of symptom onset in carriers, but not non-carriers. Regions included the bilateral insulae/orbitofrontal cortices, anterior cingulate/paracingulate gyri, and inferior parietal cortices, as well as the left middle temporal gyrus. For all bilateral regions, associations were greater on the right side. After correction for partial volume effects in a region of interest analysis, the results were found to be largely driven by the C9orf72 genetic subgroup. These cerebral blood flow differences first appeared approximately 12.5 years before the expected symptom onset determined on an individual basis. Cerebral blood flow was lower in presymptomatic mutation carriers closer to and beyond their expected age of symptom onset in key frontotemporal dementia signature regions. These results suggest that arterial spin labelling MRI may be a promising non-invasive imaging biomarker for the presymptomatic stages of genetic frontotemporal dementia.
Collapse
Affiliation(s)
- Henri J M M Mutsaerts
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Saira S Mirza
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Jan Petr
- PET Center, Institute of Radiopharmaceutical Cancer Research, Helmholtz-Zentrum Dresden-Rossendorf, Dresden, Germany
| | - David L Thomas
- Institute of Neurology, University College London, London, UK
| | - David M Cash
- Institute of Neurology, University College London, London, UK
| | | | - Enrico de Vita
- Institute of Neurology, University College London, London, UK
| | - Arron W S Metcalfe
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Zahra Shirzadi
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Andrew D Robertson
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Maria Carmela Tartaglia
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada.,Memory Clinic, University Health Network, Toronto, Canada.,Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Sara B Mitchell
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Canada.,Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada.,L.C. Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Sandra E Black
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Canada.,Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada.,L.C. Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, Canada
| | - Morris Freedman
- Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada.,Baycrest Centre for Geriatric Care, Toronto, Canada
| | - David Tang-Wai
- Memory Clinic, University Health Network, Toronto, Canada.,Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada
| | - Ron Keren
- Memory Clinic, University Health Network, Toronto, Canada
| | - Ekaterina Rogaeva
- Tanz Centre for Research in Neurodegenerative Diseases, University of Toronto, Toronto, Canada
| | - John van Swieten
- Department of Neurology, Erasmus Medical Center, Rotterdam, The Netherlands
| | - Robert Laforce
- Clinique Interdisciplinaire de Mémoire (CIME), Département des Sciences Neurologiques, CHU de Québec, Faculté de médecine, Université Laval, Québec, Canada
| | - Fabrizio Tagliavini
- Fondazione Istituto di Ricovero e Cura a Carattere Scientifico, Milan, Italy
| | - Barbara Borroni
- Department of Medical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - Daniela Galimberti
- Centro Dino Ferrari, Fondazione Ca' Granda IRCCS Ospedale Policlinico, University of Milan, Milan, Italy
| | - James B Rowe
- Department of Clinical Neurosciences, University of Cambridge, Cambridge, UK
| | - Caroline Graff
- Department of Geriatric Medicine, Karolinska Institutet, Stockholm, Sweden
| | | | - Elizabeth Finger
- Department of Clinical Neurological Sciences, University of Western Ontario, London, Canada
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | | | | | - Bradley J MacIntosh
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Canada
| | - Mario Masellis
- Hurvitz Brain Sciences Program, Sunnybrook Research Institute, University of Toronto, Toronto, Canada.,Division of Neurology, Department of Medicine, Sunnybrook Health Sciences Centre, University of Toronto, Toronto, Canada.,L.C. Campbell Cognitive Neurology Research Unit, Sunnybrook Health Sciences Centre, Toronto, Canada.,Cognitive and Movement Disorders Clinic, Sunnybrook Health Sciences Centre, Toronto, Canada
| | | |
Collapse
|
38
|
Reddy RG, Surineni G, Bhattacharya D, Marvadi SK, Sagar A, Kalle AM, Kumar A, Kantevari S, Chakravarty S. Crafting Carbazole-Based Vorinostat and Tubastatin-A-like Histone Deacetylase (HDAC) Inhibitors with Potent in Vitro and in Vivo Neuroactive Functions. ACS OMEGA 2019; 4:17279-17294. [PMID: 31656902 PMCID: PMC6811854 DOI: 10.1021/acsomega.9b01950] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 06/28/2019] [Accepted: 09/23/2019] [Indexed: 06/10/2023]
Abstract
Small-molecule inhibitors of HDACs (HDACi) induce hyperacetylation of histone and nonhistone proteins and have emerged as potential therapeutic agents in most animal models tested. The established HDACi vorinostat and tubastatin-A alleviate neurodegenerative and behavioral conditions in animal models of neuropsychiatric disorders restoring the neurotrophic milieu. In spite of the neuroactive pharmacological role of HDACi (vorinostat and tubastatin-A), they are limited by efficacy and toxicity. Considering these limitations and concern, we have designed novel compounds 3-11 as potential HDACi based on the strategic crafting of the key pharmacophoric elements of vorinostat and tubastatin-A into architecting a single molecule. The molecules 3-11 were synthesized through a multistep reaction sequence starting from carbazole and were fully characterized by NMR and mass spectral analysis. The novel molecules 3-11 showed remarkable pan HDAC inhibition and the potential to increase the levels of acetyl H3 and acetyl tubulin. In addition, few novel HDAC inhibitors 4-8, 10, and 11 exhibited significant neurite outgrowth-promoting activity with no observable cytotoxic effects, and interestingly, compound 5 has shown comparably more neurite growth than the parent compounds vorinostat and tubastatin-A. Also, compound 5 was evaluated for possible mood-elevating effects in a chronic unpredictable stress model of Zebrafish. It showed potent anxiolytic and antidepressant-like effects in the novel tank test and social interaction test, respectively. Furthermore, the potent in vitro and in vivo neuroactive compound 5 has shown selectivity for class II over class I HDACs. Our results suggest that the novel carbazole-based HDAC inhibitors, crafted with vorinostat and tubastatin-A pharmacophoric moieties, have potent neurite outgrowth activity and potential to be developed as therapeutics to treat depression and related psychiatric disorders.
Collapse
Affiliation(s)
- R. Gajendra Reddy
- Applied
Biology Division and Fluoro and Agrochemical Division, CSIR-Indian
Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, Telangana, India
- Academy
of Scientific and Innovative Research (AcSIR), Chennai 600113, India
| | - Goverdhan Surineni
- Applied
Biology Division and Fluoro and Agrochemical Division, CSIR-Indian
Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, Telangana, India
| | - Dwaipayan Bhattacharya
- Applied
Biology Division and Fluoro and Agrochemical Division, CSIR-Indian
Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, Telangana, India
| | - Sandeep Kumar Marvadi
- Applied
Biology Division and Fluoro and Agrochemical Division, CSIR-Indian
Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, Telangana, India
| | - Arpita Sagar
- Department
of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, Telangana, India
| | - Arunasree M. Kalle
- Department
of Animal Biology, School of Life Sciences, University of Hyderabad, Hyderabad 500046, Telangana, India
| | - Arvind Kumar
- CSIR-Centre
for Cellular and Molecular Biology, Habsiguda, Uppal Road, Hyderabad 500007, Telangana, India
- Academy
of Scientific and Innovative Research (AcSIR), Chennai 600113, India
| | - Srinivas Kantevari
- Applied
Biology Division and Fluoro and Agrochemical Division, CSIR-Indian
Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, Telangana, India
- Academy
of Scientific and Innovative Research (AcSIR), Chennai 600113, India
| | - Sumana Chakravarty
- Applied
Biology Division and Fluoro and Agrochemical Division, CSIR-Indian
Institute of Chemical Technology, Tarnaka, Uppal Road, Hyderabad 500007, Telangana, India
- Academy
of Scientific and Innovative Research (AcSIR), Chennai 600113, India
| |
Collapse
|
39
|
Moreno-Yruela C, Fass DM, Cheng C, Herz J, Olsen CA, Haggarty SJ. Kinetic Tuning of HDAC Inhibitors Affords Potent Inducers of Progranulin Expression. ACS Chem Neurosci 2019; 10:3769-3777. [PMID: 31330099 DOI: 10.1021/acschemneuro.9b00281] [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] [Indexed: 12/15/2022] Open
Abstract
Histone deacetylases (HDACs) are enzymes involved in the epigenetic control of gene expression. A handful of HDAC inhibitors have been approved for the treatment of cancer, and HDAC inhibition has also been proposed as a novel therapeutic strategy for neurodegenerative disorders. These disorders include progranulin (PGRN)-deficient forms of frontotemporal dementia caused by mutations in the GRN gene that lead to haploinsufficiency. Hydroxamic-acid-based inhibitors of HDACs 1-3, reported to have fast-on/fast-off binding kinetics, induce increased expression of PGRN in human neuronal models, while the benzamide class of slow-binding HDAC inhibitors does not produce this effect. These observations indicate that the kinetics of HDAC inhibitor binding can be tuned for optimal induction of human PGRN expression in neurons. Here, we further expand on these findings using human cortical-like, glutamatergic neurons. We provide evidence that two prototypical, potent hydroxamic acid HDAC inhibitors that induce PGRN (panobinostat and trichostatin A) exhibit an initial fast-binding step followed by a second, slower step, referred to as mechanism B of slow binding, rather than simpler fast-on/fast-off binding kinetics. In addition, we show that trapoxin A, a macrocyclic, epoxyketone-containing class I HDAC inhibitor, exhibits slow binding with high, picomolar potency and also induces PGRN expression in human neurons. Finally, we demonstrate induction of PGRN expression by fast-on/fast-off, highly potent, macrocyclic HDAC inhibitors with ethyl ketone or ethyl ester Zn2+ binding groups. Taken together, these data expand our understanding of HDAC1-3 inhibitor binding kinetics, and further delineate the specific combinations of structural and kinetic features of HDAC inhibitors that are optimal for upregulating PGRN expression in human neurons and thus may have translational relevance in neurodegenerative disease.
Collapse
Affiliation(s)
- Carlos Moreno-Yruela
- Center for Biopharmaceuticals & Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Departments of Neurology & Psychiatry, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, Massachusetts 02114, United States
| | - Daniel M. Fass
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Departments of Neurology & Psychiatry, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, Massachusetts 02114, United States
| | - Chialin Cheng
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Departments of Neurology & Psychiatry, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, Massachusetts 02114, United States
| | - Joachim Herz
- Departments of Molecular Genetics, Neuroscience, Neurology and Neurotherapeutics, Center for Translational Neurodegeneration Research, University of Texas Southwestern Medical Center at Dallas, Dallas, Texas 75390-9046, United States
| | - Christian A. Olsen
- Center for Biopharmaceuticals & Department of Drug Design and Pharmacology, Faculty of Health and Medical Sciences, University of Copenhagen, DK-2100 Copenhagen, Denmark
| | - Stephen J. Haggarty
- Chemical Neurobiology Laboratory, Center for Genomic Medicine, Departments of Neurology & Psychiatry, Massachusetts General Hospital and Harvard Medical School, 185 Cambridge Street, Boston, Massachusetts 02114, United States
| |
Collapse
|
40
|
Butler VJ, Gao F, Corrales CI, Cortopassi WA, Caballero B, Vohra M, Ashrafi K, Cuervo AM, Jacobson MP, Coppola G, Kao AW. Age- and stress-associated C. elegans granulins impair lysosomal function and induce a compensatory HLH-30/TFEB transcriptional response. PLoS Genet 2019; 15:e1008295. [PMID: 31398187 PMCID: PMC6703691 DOI: 10.1371/journal.pgen.1008295] [Citation(s) in RCA: 20] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 08/21/2019] [Accepted: 07/07/2019] [Indexed: 12/11/2022] Open
Abstract
The progressive failure of protein homeostasis is a hallmark of aging and a common feature in neurodegenerative disease. As the enzymes executing the final stages of autophagy, lysosomal proteases are key contributors to the maintenance of protein homeostasis with age. We previously reported that expression of granulin peptides, the cleavage products of the neurodegenerative disease protein progranulin, enhance the accumulation and toxicity of TAR DNA binding protein 43 (TDP-43) in Caenorhabditis elegans (C. elegans). In this study we show that C. elegans granulins are produced in an age- and stress-dependent manner. Granulins localize to the endolysosomal compartment where they impair lysosomal protease expression and activity. Consequently, protein homeostasis is disrupted, promoting the nuclear translocation of the lysosomal transcription factor HLH-30/TFEB, and prompting cells to activate a compensatory transcriptional program. The three C. elegans granulin peptides exhibited distinct but overlapping functional effects in our assays, which may be due to amino acid composition that results in distinct electrostatic and hydrophobicity profiles. Our results support a model in which granulin production modulates a critical transition between the normal, physiological regulation of protease activity and the impairment of lysosomal function that can occur with age and disease.
Collapse
Affiliation(s)
- Victoria J. Butler
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California, United States of America
| | - Fuying Gao
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Christian I. Corrales
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California, United States of America
| | - Wilian A. Cortopassi
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California, United States of America
| | - Benjamin Caballero
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, New York, United States of America
| | - Mihir Vohra
- Department of Physiology, University of California, San Francisco, California, United States of America
| | - Kaveh Ashrafi
- Department of Physiology, University of California, San Francisco, California, United States of America
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, New York, United States of America
| | - Matthew P. Jacobson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California, United States of America
| | - Giovanni Coppola
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Aimee W. Kao
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
41
|
Elia LP, Mason AR, Alijagic A, Finkbeiner S. Genetic Regulation of Neuronal Progranulin Reveals a Critical Role for the Autophagy-Lysosome Pathway. J Neurosci 2019; 39:3332-3344. [PMID: 30696728 PMCID: PMC6788815 DOI: 10.1523/jneurosci.3498-17.2019] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2018] [Revised: 01/16/2019] [Accepted: 01/16/2019] [Indexed: 12/29/2022] Open
Abstract
Deficient progranulin levels cause dose-dependent neurological syndromes: haploinsufficiency leads to frontotemporal lobar degeneration (FTLD) and nullizygosity produces adult-onset neuronal ceroid lipofuscinosis. Mechanisms controlling progranulin levels are largely unknown. To better understand progranulin regulation, we performed a genome-wide RNAi screen using an ELISA-based platform to discover genes that regulate progranulin levels in neurons. We identified 830 genes that raise or lower progranulin levels by at least 1.5-fold in Neuro2a cells. When inhibited by siRNA or some by submicromolar concentrations of small-molecule inhibitors, 33 genes of the druggable genome increased progranulin levels in mouse primary cortical neurons; several of these also raised progranulin levels in FTLD model mouse neurons. "Hit" genes regulated progranulin by transcriptional or posttranscriptional mechanisms. Pathway analysis revealed enrichment of hit genes from the autophagy-lysosome pathway (ALP), suggesting a key role for this pathway in regulating progranulin levels. Progranulin itself regulates lysosome function. We found progranulin deficiency in neurons increased autophagy and caused abnormally enlarged lysosomes and boosting progranulin levels restored autophagy and lysosome size to control levels. Our data link the ALP to neuronal progranulin: progranulin levels are regulated by autophagy and, in turn, progranulin regulates the ALP. Restoring progranulin levels by targeting genetic modifiers reversed FTLD functional deficits, opening up potential opportunities for future therapeutics development.SIGNIFICANCE STATEMENT Progranulin regulates neuron and immune functions and is implicated in aging. Loss of one functional allele causes haploinsufficiency and leads to frontotemporal lobar degeneration (FTLD), the second leading cause of dementia. Progranulin gene polymorphisms are linked to Alzheimer's disease (AD) and complete loss of function causes neuronal ceroid lipofuscinosis. Despite the critical role of progranulin levels in neurodegenerative disease risk, almost nothing is known about their regulation. We performed an unbiased screen and identified specific pathways controlling progranulin levels in neurons. Modulation of these pathways restored levels in progranulin-deficient neurons and reversed FTLD phenotypes. We provide a new comprehensive understanding of the genetic regulation of progranulin levels and identify potential targets to treat FTLD and other neurodegenerative diseases, including AD.
Collapse
Affiliation(s)
- Lisa P Elia
- Center for Systems and Therapeutics and Taube/Koret Center for Neurodegenerative Disease Research, San Francisco, California,
- The J. David Gladstone Institutes, San Francisco, California 94158
| | - Amanda R Mason
- Keck School of Medicine, University of Southern California, Los Angeles, California, 90033, and
| | - Amela Alijagic
- The J. David Gladstone Institutes, San Francisco, California 94158
| | - Steven Finkbeiner
- Center for Systems and Therapeutics and Taube/Koret Center for Neurodegenerative Disease Research, San Francisco, California,
- The J. David Gladstone Institutes, San Francisco, California 94158
- Departments of Neurology and Physiology, University of California, San Francisco, California 94143
| |
Collapse
|
42
|
Logroscino G, Imbimbo BP, Lozupone M, Sardone R, Capozzo R, Battista P, Zecca C, Dibello V, Giannelli G, Bellomo A, Greco A, Daniele A, Seripa D, Panza F. Promising therapies for the treatment of frontotemporal dementia clinical phenotypes: from symptomatic to disease-modifying drugs. Expert Opin Pharmacother 2019; 20:1091-1107. [PMID: 31002267 DOI: 10.1080/14656566.2019.1598377] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
INTRODUCTION Frontotemporal dementia (FTD) is a heterogeneous clinical entity that includes several disorders characterized by different cellular mechanisms. Distinctive clinical features in FTD include behavioral, affective, and cognitive symptoms. Unfortunately, little progress has been made over the past 20 years in terms of the development of effective disease-modifying drugs with the currently available symptomatic treatments having limited clinical utility. AREAS COVERED This article reviews the principal pharmacological intervention studies for FTD. These are predominantly randomized clinical trials and include symptomatic treatments and potential disease-modifying drugs. EXPERT OPINION There is insufficient evidence on effective treatments for FTD and studies with better methodological backgrounds are needed. Most studies reporting therapeutic benefits were conducted with selective serotonin reuptake inhibitors, while anti-dementia drugs have been ineffective in FTD. Since the underlying pathology of FTD mostly consists of abnormal tau protein or TDP-43 aggregates, treatments are being developed to interfere with their aggregation process or with the clearance of these proteins. Furthermore, disease-modifying treatments remain years away as demonstrated by the recent negative Phase III findings of a tau aggregation inhibitor (LMTM) for treating the behavioral variant of FTD. The results from current ongoing Phase I/II trials will hopefully give light to future treatment options.
Collapse
Affiliation(s)
- Giancarlo Logroscino
- a Neurodegenerative Disease Unit, Department of Basic Medical Sciences, Neuroscience and Sense Organs , University of Bari "Aldo Moro" , Bari , Italy.,b Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain , University of Bari "Aldo Moro", "Pia Fondazione Cardinale G. Panico" , Lecce , Italy
| | - Bruno P Imbimbo
- c Department of Research and Development , Chiesi Farmaceutici , Parma , Italy
| | - Madia Lozupone
- a Neurodegenerative Disease Unit, Department of Basic Medical Sciences, Neuroscience and Sense Organs , University of Bari "Aldo Moro" , Bari , Italy
| | - Rodolfo Sardone
- d National Institute of Gastroenterology "Saverio de Bellis" , Research Hospital , Castellana Grotte Bari , Italy
| | - Rosa Capozzo
- b Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain , University of Bari "Aldo Moro", "Pia Fondazione Cardinale G. Panico" , Lecce , Italy
| | - Petronilla Battista
- e Istituti Clinici Scientifici Maugeri SPA SB, IRCCS , Institute of Cassano Murge , Bari , Italy
| | - Chiara Zecca
- b Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain , University of Bari "Aldo Moro", "Pia Fondazione Cardinale G. Panico" , Lecce , Italy
| | - Vittorio Dibello
- d National Institute of Gastroenterology "Saverio de Bellis" , Research Hospital , Castellana Grotte Bari , Italy.,f Interdisciplinary Department of Medicine (DIM), Section of Dentistry , University of Bari AldoMoro , Bari , Italy
| | - Gianluigi Giannelli
- d National Institute of Gastroenterology "Saverio de Bellis" , Research Hospital , Castellana Grotte Bari , Italy
| | - Antonello Bellomo
- g Psychiatric Unit, Department of Clinical and Experimental Medicine , University of Foggia , Foggia , Italy
| | - Antonio Greco
- h Geriatric Unit , Fondazione IRCCS "Casa Sollievo della Sofferenza" , Foggia , Italy
| | - Antonio Daniele
- i Institute of Neurology , Catholic University of Sacred Heart , Rome , Italy.,j Institute of Neurology, Fondazione Policlinico Universitario A. Gemelli IRCCS , Rome , Italy
| | - Davide Seripa
- h Geriatric Unit , Fondazione IRCCS "Casa Sollievo della Sofferenza" , Foggia , Italy
| | - Francesco Panza
- a Neurodegenerative Disease Unit, Department of Basic Medical Sciences, Neuroscience and Sense Organs , University of Bari "Aldo Moro" , Bari , Italy.,b Department of Clinical Research in Neurology, Center for Neurodegenerative Diseases and the Aging Brain , University of Bari "Aldo Moro", "Pia Fondazione Cardinale G. Panico" , Lecce , Italy.,d National Institute of Gastroenterology "Saverio de Bellis" , Research Hospital , Castellana Grotte Bari , Italy.,h Geriatric Unit , Fondazione IRCCS "Casa Sollievo della Sofferenza" , Foggia , Italy
| |
Collapse
|
43
|
Palomo V, Tosat-Bitrian C, Nozal V, Nagaraj S, Martin-Requero A, Martinez A. TDP-43: A Key Therapeutic Target beyond Amyotrophic Lateral Sclerosis. ACS Chem Neurosci 2019; 10:1183-1196. [PMID: 30785719 DOI: 10.1021/acschemneuro.9b00026] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
Abstract
Accumulation of TDP-43 in the cytoplasm of diseased neurons is the pathological hallmark of frontotemporal dementia-TDP (FTLD-TDP) and amyotrophic lateral sclerosis (ALS), two diseases that lack efficacious medicine to prevent or to stop disease progression. The discovery of mutations in the TARDBP gene (encoding the nuclear protein known as TDP-43) in both FTLD and ALS patients provided evidence for a link between TDP-43 alterations and neurodegeneration. Our understanding of TDP-43 function has advanced profoundly in the past several years; however, its complete role and the molecular mechanisms that lead to disease are not fully understood. Here we summarize the recent studies of this protein, its relation to neurodegenerative diseases, and the therapeutic strategies for restoring its homeostasis with small molecules. Finally, we briefly discuss the available cellular and animal models that help to shed light on TDP-43 pathology and could serve as tools for the discovery of pharmacological agents for the treatment of TDP-43-related diseases.
Collapse
Affiliation(s)
- Valle Palomo
- Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, 28031 Madrid, Spain
| | | | - Vanesa Nozal
- Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Siranjeevi Nagaraj
- Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - Angeles Martin-Requero
- Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, 28031 Madrid, Spain
| | - Ana Martinez
- Centro de Investigaciones Biológicas, CSIC, Ramiro de Maeztu 9, 28040 Madrid, Spain
- Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Instituto Carlos III, 28031 Madrid, Spain
| |
Collapse
|
44
|
Pharmacologic normalization of pathogenic dosage underlying genetic diseases: an overview of the literature and path forward. Emerg Top Life Sci 2019; 3:53-62. [PMID: 33523192 DOI: 10.1042/etls20180099] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2019] [Revised: 02/22/2019] [Accepted: 02/25/2019] [Indexed: 12/17/2022]
Abstract
Most monogenic disorders are caused by a pathologic deficit or excess of a single transcript and/or protein. Given that small molecules, including drugs, can affect levels of mRNA and protein, the pharmacologic normalization of such pathogenic dosage represents a possible therapeutic approach for such conditions. Here, we review the literature exploring pharmacologic modulation of mRNA and/or protein levels for disorders with paralogous modifier genes, for haploinsufficient disorders (insufficient gene-product), as well as toxic gain-of-function disorders (surplus or pathologic gene-product). We also discuss challenges facing the development of rare disease therapy by pharmacologic modulation of mRNA and protein. Finally, we lay out guiding principles for selection of disorders which may be amenable to this approach.
Collapse
|
45
|
Lee SE, Sias AC, Kosik EL, Flagan TM, Deng J, Chu SA, Brown JA, Vidovszky AA, Ramos EM, Gorno-Tempini ML, Karydas AM, Coppola G, Geschwind DH, Rademakers R, Boeve BF, Boxer AL, Rosen HJ, Miller BL, Seeley WW. Thalamo-cortical network hyperconnectivity in preclinical progranulin mutation carriers. Neuroimage Clin 2019; 22:101751. [PMID: 30921613 PMCID: PMC6438992 DOI: 10.1016/j.nicl.2019.101751] [Citation(s) in RCA: 34] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2018] [Revised: 03/02/2019] [Accepted: 03/09/2019] [Indexed: 12/12/2022]
Abstract
Mutations in progranulin (GRN) cause heterogeneous clinical syndromes, including behavioral variant frontotemporal dementia (bvFTD), primary progressive aphasia (PPA), corticobasal syndrome (CBS) and Alzheimer-type dementia (AD-type dementia). Human studies have shown that presymptomatic GRN carriers feature reduced connectivity in the salience network, a system targeted in bvFTD. Mice with homozygous deletion of GRN, in contrast, show thalamo-cortical hypersynchrony due to aberrant pruning of inhibitory synapses onto thalamo-cortical projection neurons. No studies have systematically explored the intrinsic connectivity networks (ICNs) targeted by the four GRN-associated clinical syndromes, or have forged clear links between human and mouse model findings. We compared 17 preclinical GRN carriers (14 "presymptomatic" clinically normal and three "prodromal" with mild cognitive symptoms) to healthy controls to assess for differences in cognitive testing and gray matter volume. Using task-free fMRI, we assessed connectivity in the salience network, a non-fluent variant primary progressive aphasia network (nfvPPA), the perirolandic network (CBS), and the default mode network (AD-type dementia). GRN carriers and controls showed similar performance on cognitive testing. Although carriers showed little evidence of brain atrophy, markedly enhanced connectivity emerged in all four networks, and thalamo-cortical hyperconnectivity stood out as a unifying feature. Voxelwise assessment of whole brain degree centrality, an unbiased graph theoretical connectivity metric, confirmed thalamic hyperconnectivity. These results show that human GRN disease and the prevailing GRN mouse model share a thalamo-cortical network hypersynchrony phenotype. Longitudinal studies will determine whether this network physiology represents a compensatory response as carriers approach symptom onset, or an early and sustained preclinical manifestation of lifelong progranulin haploinsufficiency.
Collapse
Affiliation(s)
- Suzee E Lee
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States.
| | - Ana C Sias
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Eena L Kosik
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Taru M Flagan
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Jersey Deng
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Stephanie A Chu
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Jesse A Brown
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Anna A Vidovszky
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Eliana Marisa Ramos
- University of California, Neurobehavior Division, Department of Neurology, Los Angeles, United States
| | - Maria Luisa Gorno-Tempini
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Anna M Karydas
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Giovanni Coppola
- University of California, Neurobehavior Division, Department of Neurology, Los Angeles, United States
| | - Daniel H Geschwind
- University of California, Neurobehavior Division, Department of Neurology, Los Angeles, United States
| | - Rosa Rademakers
- Mayo Clinic Jacksonville, Department of Neuroscience, Jacksonville, United States
| | - Bradley F Boeve
- Mayo Clinic, Department of Neurology, Rochester, United States
| | - Adam L Boxer
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Howard J Rosen
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - Bruce L Miller
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States
| | - William W Seeley
- University of California, Memory and Aging Center, Department of Neurology, San Francisco, United States; University of California, Department of Pathology, San Francisco, United States
| |
Collapse
|
46
|
Amado DA, Rieders JM, Diatta F, Hernandez-Con P, Singer A, Mak JT, Zhang J, Lancaster E, Davidson BL, Chen-Plotkin AS. AAV-Mediated Progranulin Delivery to a Mouse Model of Progranulin Deficiency Causes T Cell-Mediated Toxicity. Mol Ther 2019; 27:465-478. [PMID: 30559071 PMCID: PMC6369714 DOI: 10.1016/j.ymthe.2018.11.013] [Citation(s) in RCA: 42] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2018] [Revised: 11/01/2018] [Accepted: 11/11/2018] [Indexed: 11/16/2022] Open
Abstract
Adeno-associated virus-mediated gene replacement is emerging as a safe and effective means of correcting single-gene mutations affecting the CNS. AAV-mediated progranulin gene (GRN) delivery has been proposed as a treatment for GRN-deficient frontotemporal dementia and neuronal ceroid lipofuscinosis, and recent studies using intraparenchymal AAV-Grn delivery to brain have shown moderate success in histopathologic and behavioral rescue in mouse models. Here, we used AAV9 to deliver GRN to the lateral ventricle to achieve widespread expression in the Grn null mouse brain. We found that, despite a global increase in progranulin, overexpression resulted in dramatic and selective hippocampal toxicity and degeneration affecting neurons and glia. Hippocampal degeneration was preceded by T cell infiltration and perivascular cuffing. GRN delivery with an ependymal-targeting AAV for selective secretion of progranulin into the cerebrospinal fluid similarly resulted in T cell infiltration, as well as ependymal hypertrophy. Interestingly, overexpression of GRN in wild-type animals also provoked T cell infiltration. These results call into question the safety of GRN overexpression in the CNS, with evidence for both a region-selective immune response and cellular proliferative response. Our results highlight the importance of careful consideration of target gene biology and cellular response to overexpression prior to progressing to the clinic.
Collapse
Affiliation(s)
- Defne A Amado
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Julianne M Rieders
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Children's Hospital of Philadelphia, 3501 Civic Center Boulevard, 5060 CTRB, Philadelphia, PA 19104, USA
| | - Fortunay Diatta
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Pilar Hernandez-Con
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Adina Singer
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Jordan T Mak
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Junxian Zhang
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Eric Lancaster
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Beverly L Davidson
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA; Children's Hospital of Philadelphia, 3501 Civic Center Boulevard, 5060 CTRB, Philadelphia, PA 19104, USA.
| | - Alice S Chen-Plotkin
- Department of Neurology, Perelman School of Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
47
|
Cui Y, Hettinghouse A, Liu CJ. Progranulin: A conductor of receptors orchestra, a chaperone of lysosomal enzymes and a therapeutic target for multiple diseases. Cytokine Growth Factor Rev 2019; 45:53-64. [PMID: 30733059 DOI: 10.1016/j.cytogfr.2019.01.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 01/29/2019] [Indexed: 12/14/2022]
Abstract
Progranulin (PGRN), a widely expressed glycoprotein with pleiotropic function, has been linked to a host of physiological processes and diverse pathological states. A series of contemporary preclinical disease models and clinical trials have evaluated various therapeutic strategies targeting PGRN, highlighting PGRN as a promising therapeutic target. Herein we summarize available knowledge of PGRN targeting in various kinds of diseases, including common neurological diseases, inflammatory autoimmune diseases, cancer, tissue repair, and rare lysosomal storage diseases, with a focus on the functional domain-oriented drug development strategies. In particular, we emphasize the role of extracellular PGRN as a non-conventional, extracellular matrix bound, growth factor-like conductor orchestrating multiple membrane receptors and intracellular PGRN as a chaperone/co-chaperone that mediates the folding and traffic of its various binding partners.
Collapse
Affiliation(s)
- Yazhou Cui
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003, USA; Shandong Medical Biotechnological Center, Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Aubryanna Hettinghouse
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003, USA
| | - Chuan-Ju Liu
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003, USA; Department of Cell Biology, New York University School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|
48
|
Abstract
Frontotemporal dementia (FTD) is a common young-onset dementia presenting with heterogeneous and distinct syndromes. It is characterized by progressive deficits in behavior, language, and executive function. The disease may exhibit similar characteristics to many psychiatric disorders owing to its prominent behavioral features. The concept of precision medicine has recently emerged, and it involves neurodegenerative disease treatment that is personalized to match an individual's specific pattern of neuroimaging, neuropathology, and genetic variability. In this paper, the pathophysiology underlying FTD, which is characterized by the selective degeneration of the frontal and temporal cortices, is reviewed. We also discuss recent advancements in FTD research from the perspectives of clinical, imaging, molecular characterizations, and treatment. This review focuses on the approach of precision medicine to manage the clinical and biological complexities of FTD.
Collapse
Affiliation(s)
- Mu-N Liu
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan.,Department of Psychiatry, Taipei Veterans General Hospital, Taipei, Taiwan.,Department of Neurology, Memory and Aging Centre, University of California, San Francisco, San Francisco, CA, United States
| | - Chi-Ieong Lau
- Department of Neurology, Shin Kong Wu Ho-Su Memorial Hospital, Taipei, Taiwan.,Applied Cognitive Neuroscience Group, Institute of Cognitive Neuroscience, University College London, London, United Kingdom.,College of Medicine, Fu-Jen Catholic University, Taipei, Taiwan
| | - Ching-Po Lin
- Institute of Brain Science, National Yang-Ming University, Taipei, Taiwan.,Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan.,Aging and Health Research Center, National Yang Ming University, Taipei, Taiwan
| |
Collapse
|
49
|
Galimberti D, Fenoglio C, Scarpini E. Progranulin as a therapeutic target for dementia. Expert Opin Ther Targets 2018; 22:579-585. [DOI: 10.1080/14728222.2018.1487951] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Affiliation(s)
- Daniela Galimberti
- Neurodegenerative Diseases Unit, University of Milan, Centro Dino Ferrari, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Chiara Fenoglio
- Neurodegenerative Diseases Unit, University of Milan, Centro Dino Ferrari, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| | - Elio Scarpini
- Neurodegenerative Diseases Unit, University of Milan, Centro Dino Ferrari, Fondazione Cà Granda, IRCCS Ospedale Maggiore Policlinico, Milan, Italy
| |
Collapse
|
50
|
Chang KH, Lee GC, Huang CC, Kuo HC, Chen CM, Hsiao YC, Hsu HC, Hsu KJ, Lin CH, Chang CW, Lee-Chen GJ, Wu YR. Genetic and functional characters of GRN p.T487I mutation in Taiwanese patients with atypical parkinsonian disorders. Parkinsonism Relat Disord 2018. [DOI: 10.1016/j.parkreldis.2018.02.045] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|