1
|
Flury A, Aljayousi L, Park HJ, Khakpour M, Mechler J, Aziz S, McGrath JD, Deme P, Sandberg C, González Ibáñez F, Braniff O, Ngo T, Smith S, Velez M, Ramirez DM, Avnon-Klein D, Murray JW, Liu J, Parent M, Mingote S, Haughey NJ, Werneburg S, Tremblay MÈ, Ayata P. A neurodegenerative cellular stress response linked to dark microglia and toxic lipid secretion. Neuron 2025; 113:554-571.e14. [PMID: 39719704 DOI: 10.1016/j.neuron.2024.11.018] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Revised: 10/22/2024] [Accepted: 11/25/2024] [Indexed: 12/26/2024]
Abstract
The brain's primary immune cells, microglia, are a leading causal cell type in Alzheimer's disease (AD). Yet, the mechanisms by which microglia can drive neurodegeneration remain unresolved. Here, we discover that a conserved stress signaling pathway, the integrated stress response (ISR), characterizes a microglia subset with neurodegenerative outcomes. Autonomous activation of ISR in microglia is sufficient to induce early features of the ultrastructurally distinct "dark microglia" linked to pathological synapse loss. In AD models, microglial ISR activation exacerbates neurodegenerative pathologies and synapse loss while its inhibition ameliorates them. Mechanistically, we present evidence that ISR activation promotes the secretion of toxic lipids by microglia, impairing neuron homeostasis and survival in vitro. Accordingly, pharmacological inhibition of ISR or lipid synthesis mitigates synapse loss in AD models. Our results demonstrate that microglial ISR activation represents a neurodegenerative phenotype, which may be sustained, at least in part, by the secretion of toxic lipids.
Collapse
Affiliation(s)
- Anna Flury
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Leen Aljayousi
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Hye-Jin Park
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | | | - Jack Mechler
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biochemistry, CUNY Graduate Center, New York, NY 10016, USA
| | - Siaresh Aziz
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Jackson D McGrath
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Michigan Medicine, Ann Arbor, MI 48105, USA
| | - Pragney Deme
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA
| | - Colby Sandberg
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C4, Canada
| | | | - Olivia Braniff
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C4, Canada
| | - Thi Ngo
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - Simira Smith
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - Matthew Velez
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - Denice Moran Ramirez
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Dvir Avnon-Klein
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - John W Murray
- Columbia Center for Human Development, Center for Stem Cell Therapies, Department of Medicine, Columbia University Vagelos College of Physicians and Surgeons, New York, NY 10032, USA
| | - Jia Liu
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA
| | - Martin Parent
- CERVO Brain Research Center, Québec City, QC G1E 1T2, Canada
| | - Susana Mingote
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA
| | - Norman J Haughey
- Department of Neurology, Johns Hopkins University School of Medicine, Baltimore, MD 21287, USA; Department of Psychiatry, Johns Hopkins University School of Medicine, Baltimore, MD, USA
| | - Sebastian Werneburg
- Department of Ophthalmology and Visual Sciences, Kellogg Eye Center, University of Michigan, Michigan Medicine, Ann Arbor, MI 48105, USA; Michigan Neuroscience Institute, Department of Molecular and Integrative Physiology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Marie-Ève Tremblay
- Division of Medical Sciences, University of Victoria, Victoria, BC V8P 5C4, Canada; Department of Molecular Medicine, Université Laval, Québec City, QC G1V 0A6, Canada; Neurology and Neurosurgery Department, McGill University, Montréal, QC H3A 2B4, Canada; Canada Department of Biochemistry and Molecular Biology, University of British Columbia, Vancouver, BC V6T 2A1, Canada; Centre for Advanced Materials and Related Technology and Institute on Aging and Lifelong Health, University of Victoria, Victoria, BC V8N 5M8, Canada
| | - Pinar Ayata
- Neuroscience Initiative, Advanced Science Research Center, The City University of New York (CUNY) Graduate Center, New York, NY 10031, USA; Graduate Program in Biology, CUNY Graduate Center, New York, NY 10016, USA; Graduate Program in Biochemistry, CUNY Graduate Center, New York, NY 10016, USA.
| |
Collapse
|
2
|
Wei Z, Iyer MR, Zhao B, Deng J, Mitchell CS. Artificial Intelligence-Assisted Comparative Analysis of the Overlapping Molecular Pathophysiology of Alzheimer's Disease, Amyotrophic Lateral Sclerosis, and Frontotemporal Dementia. Int J Mol Sci 2024; 25:13450. [PMID: 39769215 PMCID: PMC11678588 DOI: 10.3390/ijms252413450] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2024] [Revised: 11/27/2024] [Accepted: 12/12/2024] [Indexed: 01/11/2025] Open
Abstract
The overlapping molecular pathophysiology of Alzheimer's Disease (AD), Amyotrophic Lateral Sclerosis (ALS), and Frontotemporal Dementia (FTD) was analyzed using relationships from a knowledge graph of 33+ million biomedical journal articles. The unsupervised learning rank aggregation algorithm from SemNet 2.0 compared the most important amino acid, peptide, and protein (AAPP) nodes connected to AD, ALS, or FTD. FTD shared 99.9% of its nodes with ALS and AD; AD shared 64.2% of its nodes with FTD and ALS; and ALS shared 68.3% of its nodes with AD and FTD. The results were validated and mapped to functional biological processes using supervised human supervision and an external large language model. The overall percentages of mapped intersecting biological processes were as follows: inflammation and immune response, 19%; synapse and neurotransmission, 19%; cell cycle, 15%; protein aggregation, 12%; membrane regulation, 11%; stress response and regulation, 9%; and gene regulation, 4%. Once normalized for node count, biological mappings for cell cycle regulation and stress response were more prominent in the intersection of AD and FTD. Protein aggregation, gene regulation, and energetics were more prominent in the intersection of ALS and FTD. Synapse and neurotransmission, membrane regulation, and inflammation and immune response were greater at the intersection of AD and ALS. Given the extensive molecular pathophysiology overlap, small differences in regulation, genetic, or environmental factors likely shape the underlying expressed disease phenotype. The results help prioritize testable hypotheses for future clinical or experimental research.
Collapse
Affiliation(s)
- Zihan Wei
- Laboratory for Pathology Dynamics, Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Meghna R. Iyer
- Laboratory for Pathology Dynamics, Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Benjamin Zhao
- Laboratory for Pathology Dynamics, Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Jennifer Deng
- Laboratory for Pathology Dynamics, Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Cassie S. Mitchell
- Laboratory for Pathology Dynamics, Department of Biomedical Engineering, Georgia Institute of Technology & Emory University School of Medicine, Atlanta, GA 30322, USA
- Center for Machine Learning at Georgia Tech, Atlanta, GA 30332, USA
| |
Collapse
|
3
|
Wang S, Weyer MP, Hummel R, Wilken-Schmitz A, Tegeder I, Schäfer MKE. Selective neuronal expression of progranulin is sufficient to provide neuroprotective and anti-inflammatory effects after traumatic brain injury. J Neuroinflammation 2024; 21:257. [PMID: 39390556 PMCID: PMC11468377 DOI: 10.1186/s12974-024-03249-7] [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: 06/24/2024] [Accepted: 09/28/2024] [Indexed: 10/12/2024] Open
Abstract
Progranulin (PGRN), which is produced in neurons and microglia, is a neurotrophic and anti-inflammatory glycoprotein. Human loss-of-function mutations cause frontotemporal dementia, and PGRN knockout (KO) mice are a model for dementia. In addition, PGRN KO mice exhibit severe phenotypes in models of traumatic or ischemic central nervous system (CNS) disorders, including traumatic brain injury (TBI). It is unknown whether restoration of progranulin expression in neurons (and not in microglia) might be sufficient to prevent excessive TBI-evoked brain damage. To address this question, we generated mice with Nestin-Cre-driven murine PGRN expression in a PGRN KO line (PGRN-KONestinGrn) to rescue PGRN in neurons. PGRN expression analysis in primary CNS cell cultures from naïve mice and in (non-) injured brain tissue from PGRN-KONestinGrn revealed expression of PGRN in neurons but not in microglia. After experimental TBI, examination of the structural brain damage at 5 days post-injury (dpi) showed that the TBI-induced loss of brain tissue and hippocampal neurons was exacerbated in PGRN-KOGrnflfl mice (PGRN knockout with the mGrn fl-STOP-fl allele, Cre-negative), as expected, whereas the tissue damage in PGRN-KONestinGrn mice was similar to that in PGRN-WT mice. Analysis of CD68+ immunofluorescent microglia and Cd68 mRNA expression showed that excessive microglial activation was rescued in PGRN-KONestinGrn mice, and the correlation of brain injury with Cd68 expression suggested that Cd68 was a surrogate marker for excessive brain injury caused by PGRN deficiency. The results show that restoring neuronal PGRN expression was sufficient to rescue the exacerbated neuropathology of TBI caused by PGRN deficiency, even in the absence of microglial PGRN. Hence, endogenous microglial PGRN expression was not essential for the neuroprotective or anti-inflammatory effects of PGRN after TBI in this study.
Collapse
Affiliation(s)
- Sudena Wang
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1 (Bld. 505), 55131, Mainz, Germany
| | - Marc-Philipp Weyer
- Institute for Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, Theodor Stern Kai 7 | Bd 74-75, Rm 4.101a, 60590, Frankfurt am Main, Germany
| | - Regina Hummel
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1 (Bld. 505), 55131, Mainz, Germany
| | - Annett Wilken-Schmitz
- Institute for Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, Theodor Stern Kai 7 | Bd 74-75, Rm 4.101a, 60590, Frankfurt am Main, Germany
| | - Irmgard Tegeder
- Institute for Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, Theodor Stern Kai 7 | Bd 74-75, Rm 4.101a, 60590, Frankfurt am Main, Germany
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Langenbeckstraße 1 (Bld. 505), 55131, 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
|
4
|
Ek M, Nilvebrant J, Nygren PÅ, Ståhl S, Lindberg H, Löfblom J. An anti-sortilin affibody-peptide fusion inhibits sortilin-mediated progranulin degradation. Front Immunol 2024; 15:1437886. [PMID: 39185427 PMCID: PMC11342335 DOI: 10.3389/fimmu.2024.1437886] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2024] [Accepted: 07/23/2024] [Indexed: 08/27/2024] Open
Abstract
Heterozygous loss-of-function mutations in the GRN gene are a common cause of frontotemporal dementia. Such mutations lead to decreased plasma and cerebrospinal fluid levels of progranulin (PGRN), a neurotrophic factor with lysosomal functions. Sortilin is a negative regulator of extracellular PGRN levels and has shown promise as a therapeutic target for frontotemporal dementia, enabling increased extracellular PGRN levels through inhibition of sortilin-mediated PGRN degradation. Here we report the development of a high-affinity sortilin-binding affibody-peptide fusion construct capable of increasing extracellular PGRN levels in vitro. By genetic fusion of a sortilin-binding affibody generated through phage display and a peptide derived from the progranulin C-terminus, an affinity protein (A3-PGRNC15*) with 185-pM affinity for sortilin was obtained. Treating PGRN-secreting and sortilin-expressing human glioblastoma U-251 cells with the fusion protein increased extracellular PGRN levels up to 2.5-fold, with an EC50 value of 1.3 nM. Our results introduce A3-PGRNC15* as a promising new agent with therapeutic potential for the treatment of frontotemporal dementia. Furthermore, the work highlights means to increase binding affinity through synergistic contribution from two orthogonal polypeptide units.
Collapse
Affiliation(s)
| | | | | | | | | | - John Löfblom
- Department of Protein Science, School of Engineering Sciences in Chemistry, Biotechnology and Health, KTH Royal Institute of Technology, Stockholm, Sweden
| |
Collapse
|
5
|
Takahashi H, Bhagwagar S, Nies SH, Ye H, Han X, Chiasseu MT, Wang G, Mackenzie IR, Strittmatter SM. Reduced progranulin increases tau and α-synuclein inclusions and alters mouse tauopathy phenotypes via glucocerebrosidase. Nat Commun 2024; 15:1434. [PMID: 38365772 PMCID: PMC10873339 DOI: 10.1038/s41467-024-45692-3] [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: 05/16/2022] [Accepted: 02/01/2024] [Indexed: 02/18/2024] Open
Abstract
Comorbid proteinopathies are observed in many neurodegenerative disorders including Alzheimer's disease (AD), increase with age, and influence clinical outcomes, yet the mechanisms remain ill-defined. Here, we show that reduction of progranulin (PGRN), a lysosomal protein associated with TDP-43 proteinopathy, also increases tau inclusions, causes concomitant accumulation of α-synuclein and worsens mortality and disinhibited behaviors in tauopathy mice. The increased inclusions paradoxically protect against spatial memory deficit and hippocampal neurodegeneration. PGRN reduction in male tauopathy attenuates activity of β-glucocerebrosidase (GCase), a protein previously associated with synucleinopathy, while increasing glucosylceramide (GlcCer)-positive tau inclusions. In neuronal culture, GCase inhibition enhances tau aggregation induced by AD-tau. Furthermore, purified GlcCer directly promotes tau aggregation in vitro. Neurofibrillary tangles in human tauopathies are also GlcCer-immunoreactive. Thus, in addition to TDP-43, PGRN regulates tau- and synucleinopathies via GCase and GlcCer. A lysosomal PGRN-GCase pathway may be a common therapeutic target for age-related comorbid proteinopathies.
Collapse
Affiliation(s)
- Hideyuki Takahashi
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT, 06536, USA
| | - Sanaea Bhagwagar
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT, 06536, USA
- College of Arts and Sciences, University of Pennsylvania, Philadelphia, PA, USA
| | - Sarah H Nies
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT, 06536, USA
- Graduate School of Cellular and Molecular Neuroscience, University of Tübingen, D-72074, Tübingen, Germany
| | - Hongping Ye
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229, USA
| | - Xianlin Han
- Barshop Institute for Longevity and Aging Studies, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229, USA
- Department of Medicine, University of Texas Health Science Center At San Antonio, San Antonio, TX, 78229, USA
| | - Marius T Chiasseu
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT, 06536, USA
| | - Guilin Wang
- Department of Molecular Biophysics and Biochemistry, School of Medicine, Yale University, New Haven, CT, 06520, USA
| | - Ian R Mackenzie
- Department of Pathology, University of British Columbia and Vancouver General Hospital, Vancouver, BC, Canada
| | - Stephen M Strittmatter
- Cellular Neuroscience, Neurodegeneration, Repair, Departments of Neurology and of Neuroscience, Yale University School of Medicine, New Haven, CT, 06536, USA.
| |
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
|
Kaplelach AK, Fox SN, Cook AK, Hall JA, Dannemiller RS, Jaunarajs KL, Arrant AE. Regulation of extracellular progranulin in medial prefrontal cortex. Neurobiol Dis 2023; 188:106326. [PMID: 37838007 PMCID: PMC10682954 DOI: 10.1016/j.nbd.2023.106326] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2023] [Revised: 09/28/2023] [Accepted: 10/11/2023] [Indexed: 10/16/2023] Open
Abstract
Progranulin is a secreted pro-protein that has anti-inflammatory and neurotrophic effects and is necessary for maintaining lysosomal function. Mutations in progranulin (GRN) are a major cause of frontotemporal dementia. Most pathogenic GRN mutations cause progranulin haploinsufficiency, so boosting progranulin levels is a promising therapeutic strategy. Progranulin is constitutively secreted, then taken up and trafficked to lysosomes. Before being taken up from the extracellular space, progranulin interacts with receptors that may mediate anti-inflammatory and growth factor-like effects. Modifying progranulin trafficking is a viable approach to boosting progranulin, but progranulin secretion and uptake by cells in the brain is poorly understood and may involve distinct mechanisms from other parts of the body. Understanding the cell types and processes that regulate extracellular progranulin in the brain could provide insight into progranulin's mechanism of action and inform design of progranulin-boosting therapies. To address this question we used microdialysis to measure progranulin in interstitial fluid (ISF) of mouse medial prefrontal cortex (mPFC). Grn+/- mice had approximately 50% lower ISF progranulin than wild-type mice, matching the reduction of progranulin in cortical tissue. Fluorescent in situ hybridization and immunofluorescence confirmed that microglia and neurons are the major progranulin-expressing cell types in the mPFC. Studies of conditional microglial (Mg-KO) and neuronal (N-KO) Grn knockout mice revealed that loss of progranulin from either cell type results in approximately 50% reduction in ISF progranulin. LPS injection (i.p.) produced an acute increase in ISF progranulin in mPFC. Depolarizing cells with KCl increased ISF progranulin, but this response was not altered in N-KO mice, indicating progranulin secretion by non-neuronal cells. Increasing neuronal activity with picrotoxin did not increase ISF progranulin. These data indicate that microglia and neurons are the source of most ISF progranulin in mPFC, with microglia likely secreting more progranulin per cell than neurons. The acute increase in ISF progranulin after LPS treatment is consistent with a role for extracellular progranulin in regulating inflammation, and may have been driven by microglia or peripheral immune cells. Finally, these data indicate that mPFC neurons engage in constitutive progranulin secretion that is not acutely changed by neuronal activity.
Collapse
Affiliation(s)
- Azariah K Kaplelach
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Stephanie N Fox
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Anna K Cook
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Justin A Hall
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Ryan S Dannemiller
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Karen L Jaunarajs
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA
| | - Andrew E Arrant
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Evelyn F. McKnight Brain Institute, Departments of Neurology and Neurobiology, University of Alabama at Birmingham, Birmingham, AL, USA.
| |
Collapse
|
8
|
Kumar A, Su Y, Sharma M, Singh S, Kim S, Peavey JJ, Suerken CK, Lockhart SN, Whitlow CT, Craft S, Hughes TM, Deep G. MicroRNA expression in extracellular vesicles as a novel blood-based biomarker for Alzheimer's disease. Alzheimers Dement 2023; 19:4952-4966. [PMID: 37071449 PMCID: PMC11663460 DOI: 10.1002/alz.13055] [Citation(s) in RCA: 28] [Impact Index Per Article: 14.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2022] [Revised: 02/16/2023] [Accepted: 02/22/2023] [Indexed: 04/19/2023]
Abstract
INTRODUCTION Brain cell-derived small extracellular vesicles (sEVs) in blood offer unique cellular and molecular information related to the onset and progression of Alzheimer's disease (AD). We simultaneously enriched six specific sEV subtypes from the plasma and analyzed a selected panel of microRNAs (miRNAs) in older adults with/without cognitive impairment. METHODS Total sEVs were isolated from the plasma of participants with normal cognition (CN; n = 11), mild cognitive impairment (MCI; n = 11), MCI conversion to AD dementia (MCI-AD; n = 6), and AD dementia (n = 11). Various brain cell-derived sEVs (from neurons, astrocytes, microglia, oligodendrocytes, pericytes, and endothelial cells) were enriched and analyzed for specific miRNAs. RESULTS miRNAs in sEV subtypes differentially expressed in MCI, MCI-AD, and AD dementia compared to the CN group clearly distinguished dementia status, with an area under the curve (AUC) > 0.90 and correlated with the temporal cortical region thickness on magnetic resonance imaging (MRI). DISCUSSION miRNA analyses in specific sEVs could serve as a novel blood-based molecular biomarker for AD. HIGHLIGHTS Multiple brain cell-derived small extracellular vesicles (sEVs) could be isolated simultaneously from blood. MicroRNA (miRNA) expression in sEVs could detect Alzheimer's disease (AD) with high specificity and sensitivity. miRNA expression in sEVs correlated with cortical region thickness on magnetic resonance imaging (MRI). Altered expression of miRNAs in sEVCD31 and sEVPDGFRβ suggested vascular dysfunction. miRNA expression in sEVs could predict the activation state of specific brain cell types.
Collapse
Affiliation(s)
- Ashish Kumar
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Yixin Su
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Mitu Sharma
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Sangeeta Singh
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Susy Kim
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Jeremy J. Peavey
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Cynthia K. Suerken
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Samuel N. Lockhart
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Christopher T. Whitlow
- Department of Biostatistics and Data Science, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Department of Radiology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Suzanne Craft
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Timothy M. Hughes
- Department of Internal Medicine, Section on Gerontology and Geriatric Medicine, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| | - Gagan Deep
- Department of Cancer Biology, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Sticht Center for Healthy Aging and Alzheimer’s Prevention, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
- Wake Forest Baptist Comprehensive Cancer Center, Wake Forest School of Medicine, Winston-Salem, North Carolina, USA
| |
Collapse
|
9
|
Antonioni A, Raho EM, Lopriore P, Pace AP, Latino RR, Assogna M, Mancuso M, Gragnaniello D, Granieri E, Pugliatti M, Di Lorenzo F, Koch G. Frontotemporal Dementia, Where Do We Stand? A Narrative Review. Int J Mol Sci 2023; 24:11732. [PMID: 37511491 PMCID: PMC10380352 DOI: 10.3390/ijms241411732] [Citation(s) in RCA: 41] [Impact Index Per Article: 20.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2023] [Revised: 07/18/2023] [Accepted: 07/19/2023] [Indexed: 07/30/2023] Open
Abstract
Frontotemporal dementia (FTD) is a neurodegenerative disease of growing interest, since it accounts for up to 10% of middle-age-onset dementias and entails a social, economic, and emotional burden for the patients and caregivers. It is characterised by a (at least initially) selective degeneration of the frontal and/or temporal lobe, generally leading to behavioural alterations, speech disorders, and psychiatric symptoms. Despite the recent advances, given its extreme heterogeneity, an overview that can bring together all the data currently available is still lacking. Here, we aim to provide a state of the art on the pathogenesis of this disease, starting with established findings and integrating them with more recent ones. In particular, advances in the genetics field will be examined, assessing them in relation to both the clinical manifestations and histopathological findings, as well as considering the link with other diseases, such as amyotrophic lateral sclerosis (ALS). Furthermore, the current diagnostic criteria will be explored, including neuroimaging methods, nuclear medicine investigations, and biomarkers on biological fluids. Of note, the promising information provided by neurophysiological investigations, i.e., electroencephalography and non-invasive brain stimulation techniques, concerning the alterations in brain networks and neurotransmitter systems will be reviewed. Finally, current and experimental therapies will be considered.
Collapse
Affiliation(s)
- Annibale Antonioni
- Unit of Clinical Neurology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
- Doctoral Program in Translational Neurosciences and Neurotechnologies, University of Ferrara, 44121 Ferrara, Italy
| | - Emanuela Maria Raho
- Unit of Clinical Neurology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
| | - Piervito Lopriore
- Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Antonia Pia Pace
- Institute of Radiology, Department of Medicine, University of Udine, University Hospital S. Maria della Misericordia, Azienda Sanitaria-Universitaria Friuli Centrale, 33100 Udine, Italy
| | - Raffaela Rita Latino
- Complex Structure of Neurology, Emergency Department, Fondazione Istituto di Ricovero e Cura a Carattere Scientifico Casa Sollievo della Sofferenza, 71013 San Giovanni Rotondo, Italy
| | - Martina Assogna
- Centro Demenze, Policlinico Tor Vergata, University of Rome 'Tor Vergata', 00133 Rome, Italy
- Non Invasive Brain Stimulation Unit, Istituto di Ricovero e Cura a Carattere Scientifico Santa Lucia, 00179 Rome, Italy
| | - Michelangelo Mancuso
- Neurological Institute, Department of Clinical and Experimental Medicine, University of Pisa, 56126 Pisa, Italy
| | - Daniela Gragnaniello
- Nuerology Unit, Neurosciences and Rehabilitation Department, Ferrara University Hospital, 44124 Ferrara, Italy
| | - Enrico Granieri
- Unit of Clinical Neurology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
| | - Maura Pugliatti
- Unit of Clinical Neurology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
| | - Francesco Di Lorenzo
- Non Invasive Brain Stimulation Unit, Istituto di Ricovero e Cura a Carattere Scientifico Santa Lucia, 00179 Rome, Italy
| | - Giacomo Koch
- Non Invasive Brain Stimulation Unit, Istituto di Ricovero e Cura a Carattere Scientifico Santa Lucia, 00179 Rome, Italy
- Iit@Unife Center for Translational Neurophysiology, Istituto Italiano di Tecnologia, 44121 Ferrara, Italy
- Section of Human Physiology, Neurosciences and Rehabilitation Department, University of Ferrara, 44121 Ferrara, Italy
| |
Collapse
|
10
|
Zhang T, Feng T, Wu K, Guo J, Nana AL, Yang G, Seeley WW, Hu F. Progranulin deficiency results in sex-dependent alterations in microglia in response to demyelination. Acta Neuropathol 2023:10.1007/s00401-023-02578-w. [PMID: 37120788 PMCID: PMC10375542 DOI: 10.1007/s00401-023-02578-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2023] [Revised: 04/17/2023] [Accepted: 04/18/2023] [Indexed: 05/01/2023]
Abstract
Heterozygous mutations in the granulin (GRN) gene, resulting in the haploinsufficiency of the progranulin (PGRN) protein, is a leading cause of frontotemporal lobar degeneration (FTLD). Complete loss of the PGRN protein causes neuronal ceroid lipofuscinosis (NCL), a lysosomal storage disorder. Polymorphisms in the GRN gene have also been associated with several other neurodegenerative diseases, including Alzheimer's disease (AD), and Parkinson's disease (PD). PGRN deficiency has been shown to cause myelination defects previously, but how PGRN regulates myelination is unknown. Here, we report that PGRN deficiency leads to a sex-dependent myelination defect with male mice showing more severe demyelination in response to cuprizone treatment. This is accompanied by exacerbated microglial proliferation and activation in the male PGRN-deficient mice. Interestingly, both male and female PGRN-deficient mice show sustained microglial activation after cuprizone removal and a defect in remyelination. Specific ablation of PGRN in microglia results in similar sex-dependent phenotypes, confirming a microglial function of PGRN. Lipid droplets accumulate in microglia specifically in male PGRN-deficient mice. RNA-seq analysis and mitochondrial function assays reveal key differences in oxidative phosphorylation in male versus female microglia under PGRN deficiency. A significant decrease in myelination and accumulation of myelin debris and lipid droplets in microglia were found in the corpus callosum regions of FTLD patients with GRN mutations. Taken together, our data support that PGRN deficiency leads to sex-dependent alterations in microglia with subsequent myelination defects.
Collapse
Affiliation(s)
- Tingting Zhang
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, 345 Weill Hall, Ithaca, NY, 14853, USA
| | - Tuancheng Feng
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, 345 Weill Hall, Ithaca, NY, 14853, USA
| | - Kenton Wu
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, 345 Weill Hall, Ithaca, NY, 14853, USA
| | - Jennifer Guo
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, 345 Weill Hall, Ithaca, NY, 14853, USA
| | - Alissa L Nana
- Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Guang Yang
- Department of Anesthesiology, Columbia University Irving Medical Center, New York, NY, 10032, USA
| | - William W Seeley
- Department of Neurology, University of California, San Francisco, CA, 94158, USA
- Department of Pathology, University of California, San Francisco, CA, 94158, USA
| | - Fenghua Hu
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, 345 Weill Hall, Ithaca, NY, 14853, USA.
| |
Collapse
|
11
|
Braun JEA. Extracellular chaperone networks and the export of J-domain proteins. J Biol Chem 2023; 299:102840. [PMID: 36581212 PMCID: PMC9867986 DOI: 10.1016/j.jbc.2022.102840] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2022] [Revised: 12/18/2022] [Accepted: 12/20/2022] [Indexed: 12/27/2022] Open
Abstract
An extracellular network of molecular chaperones protects a diverse array of proteins that reside in or pass through extracellular spaces. Proteins in the extracellular milieu face numerous challenges that can lead to protein misfolding and aggregation. As a checkpoint for proteins that move between cells, extracellular chaperone networks are of growing clinical relevance. J-domain proteins (JDPs) are ubiquitous molecular chaperones that are known for their essential roles in a wide array of fundamental cellular processes through their regulation of heat shock protein 70s. As the largest molecular chaperone family, JDPs have long been recognized for their diverse functions within cells. Some JDPs are elegantly selective for their "client proteins," some do not discriminate among substrates and others act cooperatively on the same target. The realization that JDPs are exported through both classical and unconventional secretory pathways has fueled investigation into the roles that JDPs play in protein quality control and intercellular communication. The proposed functions of exported JDPs are diverse. Studies suggest that export of DnaJB11 enhances extracellular proteostasis, that intercellular movement of DnaJB1 or DnaJB6 enhances the proteostasis capacity in recipient cells, whereas the import of DnaJB8 increases resistance to chemotherapy in recipient cancer cells. In addition, the export of DnaJC5 and concurrent DnaJC5-dependent ejection of dysfunctional and aggregation-prone proteins are implicated in the prevention of neurodegeneration. This review provides a brief overview of the current understanding of the extracellular chaperone networks and outlines the first wave of studies describing the cellular export of JDPs.
Collapse
Affiliation(s)
- Janice E A Braun
- Department of Biochemistry and Molecular Biology, Hotchkiss Brain Institute, University of Calgary, Calgary, Alberta, Canada.
| |
Collapse
|
12
|
Elia L, Herting B, Alijagic A, Buselli C, Wong L, Morrison G, Prado MA, Paulo JA, Gygi SP, Finley D, Finkbeiner S. Frontotemporal Dementia Patient Neurons With Progranulin Deficiency Display Protein Dyshomeostasis. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2023:2023.01.18.524611. [PMID: 36712069 PMCID: PMC9882405 DOI: 10.1101/2023.01.18.524611] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Haploinsufficiency of progranulin (PGRN) causes frontotemporal dementia (FTD), a devastating neurodegenerative disease with no effective treatment. PGRN is required for efficient proteostasis, as loss of neuronal PGRN results in dysfunctional lysosomes and impaired clearance and cytoplasmic aggregation of TDP-43, a protein involved in neurodegeneration in FTD. These and other events lead to neurodegeneration and neuroinflammation. However, the detailed mechanisms leading to protein dyshomeostasis in PGRN-deficient cells remain unclear. We report here the development of human cell models of FTD with PGRN-deficiency to explore the molecular mechanisms underlying proteostasis breakdown and TDP-43 aggregation in FTD. Neurons differentiated from FTD patient induced pluripotent stem cells (iPSCs) have reduced PGRN levels, and the neurons recapitulate key disease features, including impaired lysosomal function, defective TDP-43 turnover and accumulation, neurodegeneration, and death. Proteomic analysis revealed altered levels of proteins linked to the autophagy-lysosome pathway (ALP) and the ubiquitin-proteasome system (UPS) in FTD patient neurons, providing new mechanistic insights into the link between PGRN-deficiency and disease pathobiology.
Collapse
|
13
|
Zhao X, Lin Y, Liou B, Fu W, Jian J, Fannin V, Zhang W, Setchell KDR, Grabowski GA, Sun Y, Liu CJ. PGRN deficiency exacerbates, whereas a brain penetrant PGRN derivative protects, GBA1 mutation-associated pathologies and diseases. Proc Natl Acad Sci U S A 2023; 120:e2210442120. [PMID: 36574647 PMCID: PMC9910439 DOI: 10.1073/pnas.2210442120] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 11/08/2022] [Indexed: 12/28/2022] Open
Abstract
Mutations in GBA1, encoding glucocerebrosidase (GCase), cause Gaucher disease (GD) and are also genetic risks in developing Parkinson's disease (PD). Currently, the approved therapies are only effective for directly treating visceral symptoms, but not for primary neuronopathic involvement in GD (nGD). Progranulin (PGRN), encoded by GRN, is a novel modifier of GCase, but the impact of PGRN in GBA1 mutation-associated pathologies in vivo remains unknown. Herein, Grn-/- mice crossed into Gba9v/9v mice, a Gba1 mutant line homozygous for the Gba1 D409V mutation, generating Grn-/-Gba9v/9v (PG9V) mice. PG9V mice exhibited neurobehavioral deficits, early onset, and more severe GD phenotypes compared to Grn-/- and Gba9v/9v mice. Moreover, PG9V mice also displayed PD-like phenotype. Mechanistic analysis revealed that PGRN deficiency caused severe neuroinflammation with microgliosis and astrogliosis, along with impaired autophagy associated with the Gba1 mutation. A PGRN-derived peptide, termed ND7, ameliorated the disease phenotype in GD patient fibroblasts ex vivo. Unexpectedly, ND7 penetrated the blood-brain barrier (BBB) and effectively ameliorated the nGD manifestations and PD pathology in Gba9v/null and PG9V mice. Collectively, this study not only provides the first line of in vivo but also ex vivo evidence demonstrating the crucial role of PGRN in GBA1/Gba1 mutation-related pathologies, as well as a clinically relevant mouse model for mechanistic and potential therapeutics studies for nGD and PD. Importantly, a BBB penetrant PGRN-derived biologic was developed that may provide treatment for rare lysosomal storage diseases and common neurodegenerative disorders, particularly nGD and PD.
Collapse
Affiliation(s)
- Xiangli Zhao
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY10003
| | - Yi Lin
- The Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH45229
| | - Benjamin Liou
- The Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH45229
| | - Wenyu Fu
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY10003
| | - Jinlong Jian
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY10003
| | - Venette Fannin
- The Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH45229
| | - Wujuan Zhang
- The Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH45229
| | - Kenneth D. R. Setchell
- The Division of Pathology, Cincinnati Children's Hospital Medical Center, Cincinnati, OH45229
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH45229
| | - Gregory A. Grabowski
- The Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH45229
| | - Ying Sun
- The Division of Human Genetics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH45229
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH45229
| | - Chuan-ju Liu
- Department of Orthopaedic Surgery, New York University Grossman School of Medicine, New York, NY10003
- Department of Cell Biology, New York University Grossman School of Medicine, New York, NY10016
| |
Collapse
|
14
|
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
|
15
|
Ingannato A, Bessi V, Chiari A, Salvatori D, Bagnoli S, Bedin R, Ferrari C, Sorbi S, Nacmias B. GRN Missense Variants and Familial Alzheimer's Disease: Two Case Reports. J Alzheimers Dis 2023; 96:767-775. [PMID: 37899057 DOI: 10.3233/jad-230689] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2023]
Abstract
BACKGROUND Progranulin protein (GRN) is a growth factor, encoded by the GRN (Granulin precursor) gene, involved in several functions including inflammation, wound repair, signal transduction, proliferation, and tumorigenesis. Mutations in GRN gene are usually the genetic etiology of frontotemporal dementia (FTD), but different studies reported GRN mutations in Alzheimer 's disease (AD) patients. OBJECTIVE Here, we analyzed FTD linked gene GRN in 23 patients with a clinical diagnosis of AD and a family history of AD (FAD), not carrying mutations in AD candidate genes (PSEN 1, PSEN 2, and APP). In addition, Microtubule-associated protein tau (MAPT) gene was studied too. All patients underwent an extensive neuropsychological battery. METHODS Genetic analyses were performed thought PCR assay and sequencing. Variants were annotated with ANNOVAR and allele frequency was checked on population databases. In silico prediction tools were consulted to check nonsynonymous variants and their effect on protein function and structure. The clinical data were retrospectively collected from medical records. RESULTS Genetic screening of MAPT and GRN in 23 FAD patients highlighted two rare different variants in two probands (2/23 = 8,7%) located in GRN gene: R433W (p.Arg433Trp) and C521Y (p.Cys521Tyr). The R433W and C521Y are variants with uncertain significant, that are predicted to affect GRN protein structure and function, with a possible damaging effect. CONCLUSIONS Our data provide evidence of the importance of GRN genetic analysis also in the study of familial AD.
Collapse
Affiliation(s)
- Assunta Ingannato
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Valentina Bessi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Annalisa Chiari
- U.O. Neurologia, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Davide Salvatori
- Department of Biomedical, Metabolic, and Neural Science, University of Modena and Reggio Emilia, Modena, Italy
| | - Silvia Bagnoli
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Roberta Bedin
- U.O. Neurologia, Azienda Ospedaliero Universitaria di Modena, Modena, Italy
| | - Camilla Ferrari
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
| | - Sandro Sorbi
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| | - Benedetta Nacmias
- Department of Neuroscience, Psychology, Drug Research and Child Health, University of Florence, Florence, Italy
- IRCCS Fondazione Don Carlo Gnocchi, Florence, Italy
| |
Collapse
|
16
|
Novel CSF Biomarkers Tracking Autoimmune Inflammatory and Neurodegenerative Aspects of CNS Diseases. Diagnostics (Basel) 2022; 13:diagnostics13010073. [PMID: 36611365 PMCID: PMC9818715 DOI: 10.3390/diagnostics13010073] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2022] [Revised: 12/13/2022] [Accepted: 12/20/2022] [Indexed: 12/29/2022] Open
Abstract
The accurate diagnosis of neuroinflammatory (NIDs) and neurodegenerative (NDDs) diseases and the stratification of patients into disease subgroups with distinct disease-related characteristics that reflect the underlying pathology represents an unmet clinical need that is of particular interest in the era of emerging disease-modifying therapies (DMT). Proper patient selection for clinical trials and identifying those in the prodromal stages of the diseases or those at high risk will pave the way for precision medicine approaches and halt neuroinflammation and/or neurodegeneration in early stages where this is possible. Towards this direction, novel cerebrospinal fluid (CSF) biomarker candidates were developed to reflect the diseased organ's pathology better. Μisfolded protein accumulation, microglial activation, synaptic dysfunction, and finally, neuronal death are some of the pathophysiological aspects captured by these biomarkers to support proper diagnosis and screening. We also describe advances in the field of molecular biomarkers, including miRNAs and extracellular nucleic acids known as cell-free DNA and mitochondrial DNA molecules. Here we review the most important of these novel CSF biomarkers of NIDs and NDDs, focusing on their involvement in disease development and emphasizing their ability to define homogeneous disease phenotypes and track potential treatment outcomes that can be mirrored in the CSF compartment.
Collapse
|
17
|
Piehl N, van Olst L, Ramakrishnan A, Teregulova V, Simonton B, Zhang Z, Tapp E, Channappa D, Oh H, Losada PM, Rutledge J, Trelle AN, Mormino EC, Elahi F, Galasko DR, Henderson VW, Wagner AD, Wyss-Coray T, Gate D. Cerebrospinal fluid immune dysregulation during healthy brain aging and cognitive impairment. Cell 2022; 185:5028-5039.e13. [PMID: 36516855 PMCID: PMC9815831 DOI: 10.1016/j.cell.2022.11.019] [Citation(s) in RCA: 81] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2022] [Revised: 09/27/2022] [Accepted: 11/17/2022] [Indexed: 12/15/2022]
Abstract
Cerebrospinal fluid (CSF) contains a tightly regulated immune system. However, knowledge is lacking about how CSF immunity is altered with aging or neurodegenerative disease. Here, we performed single-cell RNA sequencing on CSF from 45 cognitively normal subjects ranging from 54 to 82 years old. We uncovered an upregulation of lipid transport genes in monocytes with age. We then compared this cohort with 14 cognitively impaired subjects. In cognitively impaired subjects, downregulation of lipid transport genes in monocytes occurred concomitantly with altered cytokine signaling to CD8 T cells. Clonal CD8 T effector memory cells upregulated C-X-C motif chemokine receptor 6 (CXCR6) in cognitively impaired subjects. The CXCR6 ligand, C-X-C motif chemokine ligand 16 (CXCL16), was elevated in the CSF of cognitively impaired subjects, suggesting CXCL16-CXCR6 signaling as a mechanism for antigen-specific T cell entry into the brain. Cumulatively, these results reveal cerebrospinal fluid immune dysregulation during healthy brain aging and cognitive impairment.
Collapse
Affiliation(s)
- Natalie Piehl
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Lynn van Olst
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Abhirami Ramakrishnan
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Victoria Teregulova
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Brooke Simonton
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Ziyang Zhang
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA
| | - Emma Tapp
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Divya Channappa
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Hamilton Oh
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; Graduate Program in Stem Cell and Regenerative Medicine, Stanford University, Stanford, CA, USA
| | - Patricia M Losada
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA
| | - Jarod Rutledge
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; Department of Genetics, Stanford University, Stanford, CA, USA
| | | | - Elizabeth C Mormino
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA; Department of Psychology, Stanford University, Stanford, CA, USA
| | - Fanny Elahi
- Departments of Neurology and Neuroscience, Friedman Brain Institute, Icahn School of Medicine at Mount Sinai, La Jolla, CA, USA
| | - Douglas R Galasko
- Department of Neurosciences, University of California at San Diego, La Jolla, CA, USA
| | - Victor W Henderson
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA
| | - Anthony D Wagner
- Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA; Department of Psychology, Stanford University, Stanford, CA, USA
| | - Tony Wyss-Coray
- Department of Neurology and Neurological Sciences, Stanford University School of Medicine, Stanford, CA, USA; Wu Tsai Neurosciences Institute, Stanford University, Stanford, CA, USA; Paul F. Glenn Center for the Biology of Aging, Stanford University School of Medicine, Stanford, CA, USA; The Phil and Penny Initiative for Brain Resilience, Stanford University, Stanford, CA, USA; Chemistry, Engineering, and Medicine for Human Health (ChEM-H), Stanford University, Stanford, CA, USA
| | - David Gate
- The Ken & Ruth Davee Department of Neurology, Northwestern University Feinberg School of Medicine, Chicago, IL, USA.
| |
Collapse
|
18
|
Vardarajan BN, Reyes‐Dumeyer D, Piriz AL, Lantigua RA, Medrano M, Rivera D, Jiménez‐Velázquez IZ, Martin E, Pericak‐Vance MA, Bush W, Farrer L, Haines JL, Wang L, Leung YY, Schellenberg G, Kukull W, De Jager P, Bennett DA, Schneider JA, Mayeux R. Progranulin mutations in clinical and neuropathological Alzheimer's disease. Alzheimers Dement 2022; 18:2458-2467. [PMID: 35258170 PMCID: PMC9360185 DOI: 10.1002/alz.12567] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/13/2021] [Revised: 09/07/2021] [Accepted: 12/10/2021] [Indexed: 01/31/2023]
Abstract
INTRODUCTION Progranulin (GRN) mutations occur in frontotemporal lobar degeneration (FTLD) and in Alzheimer's disease (AD), often with TDP-43 pathology. METHODS We determined the frequency of rs5848 and rare, pathogenic GRN mutations in two autopsy and one family cohort. We compared Braak stage, β-amyloid load, hyperphosphorylated tau (PHFtau) tangle density and TDP-43 pathology in GRN carriers and non-carriers. RESULTS Pathogenic GRN mutations were more frequent in all cohorts compared to the Genome Aggregation Database (gnomAD), but there was no evidence for association with AD. Pathogenic GRN carriers had significantly higher PHFtau tangle density adjusting for age, sex and APOE ε4 genotype. AD patients with rs5848 had higher frequencies of hippocampal sclerosis and TDP-43 deposits. Twenty-two rare, pathogenic GRN variants were observed in the family cohort. DISCUSSION GRN mutations in clinical and neuropathological AD increase the burden of tau-related brain pathology but show no specific association with β-amyloid load or AD.
Collapse
Affiliation(s)
- Badri N. Vardarajan
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- The Gertrude H. Sergievsky CenterCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of NeurologyCollege of Physicians and SurgeonsColumbia University and the New York Presbyterian HospitalNew YorkNew YorkUSA
| | - Dolly Reyes‐Dumeyer
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- The Gertrude H. Sergievsky CenterCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of NeurologyCollege of Physicians and SurgeonsColumbia University and the New York Presbyterian HospitalNew YorkNew YorkUSA
| | - Angel L. Piriz
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- The Gertrude H. Sergievsky CenterCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| | - Rafael A. Lantigua
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of MedicineCollege of Physicians and SurgeonsColumbia University, and the New York Presbyterian HospitalNew YorkNew YorkUSA
| | - Martin Medrano
- School of MedicinePontificia Universidad Catolica Madre y Maestra (PUCMM)SantiagoDominican Republic
| | - Diones Rivera
- Department of NeurologyCEDIMAT, Plaza de la SaludSanto DomingoDominican Republic
- School of MedicineUniversidad Pedro Henriquez Urena (UNPHU)Santo DomingoDominican Republic
| | | | - Eden Martin
- The John P. Hussman Institute for Human Genomicsand Dr. John T. Macdonald Foundation Department of Human GeneticsMiamiFloridaUSA
| | - Margaret A. Pericak‐Vance
- The John P. Hussman Institute for Human Genomicsand Dr. John T. Macdonald Foundation Department of Human GeneticsMiamiFloridaUSA
| | - William Bush
- Department of Biostatistics and EpidemiologyCase Western Reserve UniversityClevelandOhioUSA
| | - Lindsay Farrer
- Boston University School of MedicineBostonMassachusettsUSA
| | - Jonathan L. Haines
- Department of Biostatistics and EpidemiologyCase Western Reserve UniversityClevelandOhioUSA
| | - Li‐San Wang
- School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | - Yuk Yee Leung
- School of MedicineUniversity of PennsylvaniaPhiladelphiaPAUSA
| | | | - Walter Kukull
- Department of EpidemiologySchool of Public HealthUniversity of WashingtonSeattleWashingtonUSA
| | - Philip De Jager
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of NeurologyCollege of Physicians and SurgeonsColumbia University and the New York Presbyterian HospitalNew YorkNew YorkUSA
| | - David A. Bennett
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | - Julie A. Schneider
- Rush Alzheimer's Disease CenterRush University Medical CenterChicagoIllinoisUSA
| | | | - Richard Mayeux
- Taub Institute for Research on Alzheimer's Disease and the Aging BrainCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- The Gertrude H. Sergievsky CenterCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
- Department of NeurologyCollege of Physicians and SurgeonsColumbia University and the New York Presbyterian HospitalNew YorkNew YorkUSA
- Department of EpidemiologySchool of Public HealthUniversity of WashingtonSeattleWashingtonUSA
- Department of PsychiatryCollege of Physicians and SurgeonsColumbia UniversityNew YorkNew YorkUSA
| |
Collapse
|
19
|
Yang Y, Feng N, Liang L, Jiang R, Pan Y, Geng N, Fan M, Li X, Guo F. Progranulin, a moderator of estrogen/estrogen receptor α binding, regulates bone homeostasis through PERK/p-eIF2 signaling pathway. J Mol Med (Berl) 2022; 100:1191-1207. [PMID: 35838759 DOI: 10.1007/s00109-022-02233-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2022] [Revised: 06/08/2022] [Accepted: 06/29/2022] [Indexed: 11/28/2022]
Abstract
Under normal conditions, the human body employs the synergistic action of osteoblasts and osteoclasts to maintain a dynamic balance between bone formation and resorption. Bone homeostasis plays a very important role in the process of bone formation. Various bone diseases can occur if bone homeostasis is disrupted. In this study, the serum estrogen levels were significantly increased in the granulin (GRN)-deficient mice and PGRN regulates the binding of estrogen and estrogen receptor α (ERα) and then affects estrogen's ability to regulate bone formation and resorption. In addition, this study also explored the role that PGRN plays in regulating bone homeostasis by affecting the binding of estrogen and estrogen receptors through the protein kinase R-like endoplasmic reticulum kinase/phosphorylation of the eukaryotic initiation factor 2 signaling pathway. In summary, we confirmed the important role of PGRN in regulating the estrogen (E2)/ERα signal in maintaining bone homeostasis. Our findings may provide a new strategy for the treatment of osteoporosis and maintaining bone homeostasis. KEY MESSAGES: PGRN is a molecular regulator of the binding of E2 and ERα signal in maintaining bone homeostasis. PGRN plays in regulating bone homeostasis through the PERK/p-eIF2α signaling pathway. The best therapeutic effect of PGRN in osteoporosis is associated with different concentration of E2.
Collapse
Affiliation(s)
- Yuyou Yang
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing, 400016, China
| | - Naibo Feng
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing, 400016, China
| | - Li Liang
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing, 400016, China
| | - Rong Jiang
- Laboratory of Stem Cells and Tissue Engineering, Chongqing Medical University, Chongqing, 400016, China
| | - Yiming Pan
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing, 400016, China
| | - Nana Geng
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing, 400016, China
| | - Mengtian Fan
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing, 400016, China
| | - Xiaoli Li
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing, 400016, China
| | - Fengjin Guo
- Department of Cell Biology and Genetics, Core Facility of Development Biology, Chongqing Medical University, Chongqing, 400016, China.
| |
Collapse
|
20
|
Dedert C, Mishra V, Aggarwal G, Nguyen AD, Xu F. Progranulin Preserves Autophagy Flux and Mitochondrial Function in Rat Cortical Neurons Under High Glucose Stress. Front Cell Neurosci 2022; 16:874258. [PMID: 35880011 PMCID: PMC9308004 DOI: 10.3389/fncel.2022.874258] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2022] [Accepted: 06/17/2022] [Indexed: 12/02/2022] Open
Abstract
Chronic hyperglycemia in type II diabetes results in impaired autophagy function, accumulation of protein aggregates, and neurodegeneration. However, little is known about how to preserve autophagy function under hyperglycemic conditions. In this study, we tested whether progranulin (PGRN), a neurotrophic factor required for proper lysosome function, can restore autophagy function in neurons under high-glucose stress. We cultured primary cortical neurons derived from E18 Sprague-Dawley rat pups to maturity at 10 days in vitro (DIV) before incubation in high glucose medium and PGRN for 24-72 h before testing for autophagy flux, protein turnover, and mitochondrial function. We found that although PGRN by itself did not upregulate autophagy, it attenuated impairments in autophagy seen under high-glucose conditions. Additionally, buildup of the autophagosome marker light chain 3B (LC3B) and lysosome marker lysosome-associated membrane protein 2A (LAMP2A) changed in both neurons and astrocytes, indicating a possible role for glia in autophagy flux. Protein turnover, assessed by remaining advanced glycation end-product levels after a 6-h incubation, was preserved with PGRN treatment. Mitochondrial activity differed by complex, although PGRN appeared to increase overall activity in high glucose. We also found that activation of extracellular signal-regulated kinase 1/2 (ERK1/2) and glycogen synthase kinase 3β (GSK3β), kinases implicated in autophagy function, increased with PGRN treatment under stress. Together, our data suggest that PGRN prevents hyperglycemia-induced decreases in autophagy by increasing autophagy flux via increased ERK1/2 kinase activity in primary rat cortical neurons.
Collapse
Affiliation(s)
- Cass Dedert
- Department of Biology, College of Arts and Sciences, Saint Louis University, St. Louis, MO, United States
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, United States
| | - Vandana Mishra
- Department of Biology, College of Arts and Sciences, Saint Louis University, St. Louis, MO, United States
| | - Geetika Aggarwal
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, United States
- Department of Pharmacology and Physiology, School of Medicine, Saint Louis University, St. Louis, MO, United States
- Department of Internal Medicine, Division of Geriatric Medicine, School of Medicine, Saint Louis University, St. Louis, MO, United States
| | - Andrew D. Nguyen
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, United States
- Department of Pharmacology and Physiology, School of Medicine, Saint Louis University, St. Louis, MO, United States
- Department of Internal Medicine, Division of Geriatric Medicine, School of Medicine, Saint Louis University, St. Louis, MO, United States
| | - Fenglian Xu
- Department of Biology, College of Arts and Sciences, Saint Louis University, St. Louis, MO, United States
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, MO, United States
- Department of Pharmacology and Physiology, School of Medicine, Saint Louis University, St. Louis, MO, United States
- *Correspondence: Fenglian Xu,
| |
Collapse
|
21
|
Sirkis DW, Bonham LW, Johnson TP, La Joie R, Yokoyama JS. Dissecting the clinical heterogeneity of early-onset Alzheimer's disease. Mol Psychiatry 2022; 27:2674-2688. [PMID: 35393555 PMCID: PMC9156414 DOI: 10.1038/s41380-022-01531-9] [Citation(s) in RCA: 61] [Impact Index Per Article: 20.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/11/2021] [Revised: 03/07/2022] [Accepted: 03/16/2022] [Indexed: 12/14/2022]
Abstract
Early-onset Alzheimer's disease (EOAD) is a rare but particularly devastating form of AD. Though notable for its high degree of clinical heterogeneity, EOAD is defined by the same neuropathological hallmarks underlying the more common, late-onset form of AD. In this review, we describe the various clinical syndromes associated with EOAD, including the typical amnestic phenotype as well as atypical variants affecting visuospatial, language, executive, behavioral, and motor functions. We go on to highlight advances in fluid biomarker research and describe how molecular, structural, and functional neuroimaging can be used not only to improve EOAD diagnostic acumen but also enhance our understanding of fundamental pathobiological changes occurring years (and even decades) before the onset of symptoms. In addition, we discuss genetic variation underlying EOAD, including pathogenic variants responsible for the well-known mendelian forms of EOAD as well as variants that may increase risk for the much more common forms of EOAD that are either considered to be sporadic or lack a clear autosomal-dominant inheritance pattern. Intriguingly, specific pathogenic variants in PRNP and MAPT-genes which are more commonly associated with other neurodegenerative diseases-may provide unexpectedly important insights into the formation of AD tau pathology. Genetic analysis of the atypical clinical syndromes associated with EOAD will continue to be challenging given their rarity, but integration of fluid biomarker data, multimodal imaging, and various 'omics techniques and their application to the study of large, multicenter cohorts will enable future discoveries of fundamental mechanisms underlying the development of EOAD and its varied clinical presentations.
Collapse
Affiliation(s)
- Daniel W Sirkis
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Luke W Bonham
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Taylor P Johnson
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Renaud La Joie
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA
| | - Jennifer S Yokoyama
- Memory and Aging Center, Department of Neurology, Weill Institute for Neurosciences, University of California, San Francisco, San Francisco, CA, 94158, USA.
- Department of Radiology and Biomedical Imaging, University of California, San Francisco, San Francisco, CA, 94158, USA.
| |
Collapse
|
22
|
Impairment of the autophagy-lysosomal pathway in Alzheimer's diseases: Pathogenic mechanisms and therapeutic potential. Acta Pharm Sin B 2022; 12:1019-1040. [PMID: 35530153 PMCID: PMC9069408 DOI: 10.1016/j.apsb.2022.01.008] [Citation(s) in RCA: 102] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2021] [Revised: 11/09/2021] [Accepted: 12/16/2021] [Indexed: 01/18/2023] Open
Abstract
Alzheimer's disease (AD), the most common neurodegenerative disorder, is characterized by memory loss and cognitive dysfunction. The accumulation of misfolded protein aggregates including amyloid beta (Aβ) peptides and microtubule associated protein tau (MAPT/tau) in neuronal cells are hallmarks of AD. So far, the exact underlying mechanisms for the aetiologies of AD have not been fully understood and the effective treatment for AD is limited. Autophagy is an evolutionarily conserved cellular catabolic process by which damaged cellular organelles and protein aggregates are degraded via lysosomes. Recently, there is accumulating evidence linking the impairment of the autophagy–lysosomal pathway with AD pathogenesis. Interestingly, the enhancement of autophagy to remove protein aggregates has been proposed as a promising therapeutic strategy for AD. Here, we first summarize the recent genetic, pathological and experimental studies regarding the impairment of the autophagy–lysosomal pathway in AD. We then describe the interplay between the autophagy–lysosomal pathway and two pathological proteins, Aβ and MAPT/tau, in AD. Finally, we discuss potential therapeutic strategies and small molecules that target the autophagy–lysosomal pathway for AD treatment both in animal models and in clinical trials. Overall, this article highlights the pivotal functions of the autophagy–lysosomal pathway in AD pathogenesis and potential druggable targets in the autophagy–lysosomal pathway for AD treatment.
Collapse
|
23
|
Giudici KV, Guyonnet S, Morley JE, Nguyen AD, Aggarwal G, Parini A, Li Y, Bateman RJ, Vellas B, de Souto Barreto P. Interactions Between Weight Loss and Plasma Neurodegenerative Markers for Determining Cognitive Decline Among Community-Dwelling Older Adults. J Gerontol A Biol Sci Med Sci 2022; 77:1159-1168. [PMID: 35034116 PMCID: PMC9159663 DOI: 10.1093/gerona/glac015] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2021] [Indexed: 01/18/2023] Open
Abstract
This study aimed to investigate the interaction between weight loss (WL) and plasma amyloid-β 42/40 (Aβ 42/40), neurofilament light chain (NfL), progranulin, and their association with cognitive decline over time among older adults. This 5-year observational approach included 470 participants from the Multidomain Alzheimer Preventive Trial, mean age 76.8 years (SD = 4.5), 59.4% women. WL was defined as ≥5% decrease over the first year. Biomarkers were measured at 12 months. Cognitive function was assessed yearly from 12 months onward by Mini-Mental State Examination (MMSE); Clinical Dementia Rating sum of boxes (CDR-SB); a composite score based on Category Naming Test; Digit Symbol Substitution Test; 10 MMSE orientation items (MMSEO) and free and total recall of the Free and Cued Selective Reminding test; and these tests individually. Twenty-seven participants (5.7%) presented WL. In adjusted analyses, combined WL + lower Aβ 42/40 (≤0.103, lowest quartile) was related with more pronounced 4-year cognitive decline according to CDR-SB (p < .0001) and MMSEO (p = .021), compared with non-WL + higher Aβ 42/40. WL + higher NfL (>94.55 pg/mL, highest quartile) or progranulin (>38.4 ng/mL, 3 higher quartiles) were related with higher cognitive decline according to CDR-SB, MMSE, MMSEO, and composite score (all p < .03), compared with non-WL + lower NfL or higher progranulin. Regrouping progranulin quartiles (Q1-Q3 vs Q4) revealed higher cognitive decline among the WL + lower progranulin group compared with non-WL + lower progranulin. In conclusion, 1-year WL was associated with subsequent higher 4-year cognitive decline among older adults presenting low Aβ 42/40 or high NfL. Future studies combining plasma biomarker assessments and body weight surveillance may be useful for identifying people at risk of cognitive impairment. Clinical trial number: NCT00672685.
Collapse
Affiliation(s)
- Kelly Virecoulon Giudici
- Address correspondence to: Kelly Virecoulon Giudici, PhD, Gérontopôle of Toulouse, Institute of Aging, Toulouse University Hospital, Université Toulouse III Paul Sabatier, 37 Allée Jules Guesde, 31000 Toulouse, France. E-mail:
| | - Sophie Guyonnet
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), Toulouse, France,CERPOP UMR1295, University of Toulouse III, INSERM, UPS, Toulouse, France
| | - John E Morley
- Division of Geriatric Medicine, School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Andrew D Nguyen
- Division of Geriatric Medicine, School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Geetika Aggarwal
- Division of Geriatric Medicine, School of Medicine, Saint Louis University, St. Louis, Missouri, USA
| | - Angelo Parini
- Institute of Metabolic and Cardiovascular Diseases (I2MC), INSERM UMR 1048, University of Toulouse III Paul Sabatier, Toulouse, France
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA,Division of Biostatistics, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri, USA
| | - Bruno Vellas
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), Toulouse, France,CERPOP UMR1295, University of Toulouse III, INSERM, UPS, Toulouse, France
| | - Philipe de Souto Barreto
- Gerontopole of Toulouse, Institute of Ageing, Toulouse University Hospital (CHU Toulouse), Toulouse, France,CERPOP UMR1295, University of Toulouse III, INSERM, UPS, Toulouse, France
| | | |
Collapse
|
24
|
Zhao X, Hasan S, Liou B, Lin Y, Sun Y, Liu C. Analysis of the Biomarkers for Neurodegenerative Diseases in Aged Progranulin Deficient Mice. Int J Mol Sci 2022; 23:629. [PMID: 35054815 PMCID: PMC8775568 DOI: 10.3390/ijms23020629] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 12/23/2021] [Accepted: 01/03/2022] [Indexed: 11/28/2022] Open
Abstract
Neurodegenerative diseases are debilitating impairments that affect millions of people worldwide and are characterized by progressive degeneration of structure and function of the central or peripheral nervous system. Effective biomarkers for neurodegenerative diseases can be used to improve the diagnostic workup in the clinic as well as facilitate the development of effective disease-modifying therapies. Progranulin (PGRN) has been reported to be involved in various neurodegenerative disorders. Hence, in the current study we systematically compared the inflammation and accumulation of typical neurodegenerative disease markers in the brain tissue between PGRN knockout (PGRN KO) and wildtype (WT) mice. We found that PGRN deficiency led to significant neuron loss as well as activation of microglia and astrocytes in aged mice. Several characteristic neurodegenerative markers, including α-synuclein, TAR DNA-binding protein 43 (TDP-43), Tau, and β-amyloid, were all accumulated in the brain of PGRN-deficient mice as compared to WT mice. Moreover, higher aggregation of lipofuscin was observed in the brain tissue of PGRN-deficient mice compared with WT mice. In addition, the autophagy was also defective in the brain of PGRN-deficient mice, indicated by the abnormal expression level of autophagy marker LC3-II. Collectively, comprehensive assays support the idea that PGRN plays an important role during the development of neurodegenerative disease, indicating that PGRN might be a useful biomarker for neurodegenerative diseases in clinical settings.
Collapse
Affiliation(s)
- Xiangli Zhao
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY 10003, USA; (X.Z.); (S.H.)
| | - Sadaf Hasan
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY 10003, USA; (X.Z.); (S.H.)
| | - Benjamin Liou
- The Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (B.L.); (Y.L.); (Y.S.)
| | - Yi Lin
- The Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (B.L.); (Y.L.); (Y.S.)
| | - Ying Sun
- The Division of Human Genetics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH 45229, USA; (B.L.); (Y.L.); (Y.S.)
- Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH 45229, USA
| | - Chuanju Liu
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY 10003, USA; (X.Z.); (S.H.)
- Department of Cell Biology, New York University School of Medicine, New York, NY 10016, USA
| |
Collapse
|
25
|
Dong T, Tejwani L, Jung Y, Kokubu H, Luttik K, Driessen TM, Lim J. Microglia regulate brain progranulin levels through the endocytosis/lysosomal pathway. JCI Insight 2021; 6:e136147. [PMID: 34618685 PMCID: PMC8663778 DOI: 10.1172/jci.insight.136147] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2020] [Accepted: 10/06/2021] [Indexed: 01/01/2023] Open
Abstract
Genetic variants in Granulin (GRN), which encodes the secreted glycoprotein progranulin (PGRN), are associated with several neurodegenerative diseases, including frontotemporal lobar degeneration, neuronal ceroid lipofuscinosis, and Alzheimer's disease. These genetic alterations manifest in pathological changes due to a reduction of PGRN expression; therefore, identifying factors that can modulate PGRN levels in vivo would enhance our understanding of PGRN in neurodegeneration and could reveal novel potential therapeutic targets. Here, we report that modulation of the endocytosis/lysosomal pathway via reduction of Nemo-like kinase (Nlk) in microglia, but not in neurons, can alter total brain Pgrn levels in mice. We demonstrate that Nlk reduction promotes Pgrn degradation by enhancing its trafficking through the endocytosis/lysosomal pathway, specifically in microglia. Furthermore, genetic interaction studies in mice showed that Nlk heterozygosity in Grn haploinsufficient mice further reduces Pgrn levels and induces neuropathological phenotypes associated with PGRN deficiency. Our results reveal a mechanism for Pgrn level regulation in the brain through the active catabolism by microglia and provide insights into the pathophysiology of PGRN-associated diseases.
Collapse
Affiliation(s)
- Tingting Dong
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Leon Tejwani
- Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut, USA
- Department of Neuroscience
| | - Youngseob Jung
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Hiroshi Kokubu
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Kimberly Luttik
- Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut, USA
- Department of Neuroscience
| | - Terri M. Driessen
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
| | - Janghoo Lim
- Department of Genetics, Yale School of Medicine, New Haven, Connecticut, USA
- Interdepartmental Neuroscience Program, Yale University, New Haven, Connecticut, USA
- Department of Neuroscience
- Program in Cellular Neuroscience, Neurodegeneration and Repair, and
- Yale Stem Cell Center, Yale School of Medicine, New Haven, Connecticut, USA
| |
Collapse
|
26
|
He L, de Souto Barreto P, Giudici KV, Aggarwal G, Nguyen AD, Morley JE, Li Y, Bateman RJ, Vellas B. Cross-Sectional and Longitudinal Associations Between Plasma Neurodegenerative Biomarkers and Physical Performance Among Community-Dwelling Older Adults. J Gerontol A Biol Sci Med Sci 2021; 76:1874-1881. [PMID: 33186456 DOI: 10.1093/gerona/glaa284] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2020] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Plasma amyloid-beta (Aβ), neurofilament light chain (NfL), and progranulin (PGRN) have been related to multiple neurodegenerative conditions that might affect physical performance. The aim of this study was to explore the relationship between these plasma neurodegenerative markers and physical performance among community-dwelling older adults. METHODS Five hundred and seven older adults (aged 76 ± 5 years) previously recruited in the Multidomain Alzheimer's Preventive Trial, and had received blood and physical performance tests, were included in this study. Plasma Aβ (Aβ 42/Aβ 40 ratio), NfL, and PGRN levels were measured. Physical performance was assessed by handgrip strength and the Short Physical Performance Battery (combining gait speed, chair stands, and balance tests). Physical performance measured at the same time point and after the blood tests were used. Mixed-effect linear models were performed with age, sex, allocation to Multidomain Alzheimer's Preventive Trial group, body mass index, and Mini-Mental State Examination score as covariates. RESULTS The mean values of Aβ 42/Aβ 40 ratio, NfL, and PGRN were 0.11, 84.06 pg/mL, and 45.43 ng/mL, respectively. At the cross-sectional level, higher plasma NfL was associated with a lower Short Physical Performance Battery score (β = -0.004, 95% CI [-0.007, -0.001]). At the longitudinal level, higher PGRN levels were associated with decreasing handgrip strength over time (β = -0.02, 95% CI [-0.04, -0.007]). All the other associations were statistically nonsignificant. CONCLUSION Our findings suggest the possibility of using plasma NfL and PGRN as markers of physical performance in older adults.
Collapse
Affiliation(s)
- Lingxiao He
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, France
| | - Philipe de Souto Barreto
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, France.,UPS/Inserm UMR1027, University of Toulouse III, Toulouse, France
| | - Kelly V Giudici
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, France
| | - Geetika Aggarwal
- Division of Geriatric Medicine, Saint Louis University School of Medicine, Missouri.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Missouri
| | - Andrew D Nguyen
- Division of Geriatric Medicine, Saint Louis University School of Medicine, Missouri.,Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, Missouri
| | - John E Morley
- Division of Geriatric Medicine, Saint Louis University School of Medicine, Missouri
| | - Yan Li
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Randall J Bateman
- Department of Neurology, Washington University School of Medicine, St. Louis, Missouri
| | - Bruno Vellas
- Gérontopôle de Toulouse, Institut du Vieillissement, Centre Hospitalo-Universitaire de Toulouse, France.,UPS/Inserm UMR1027, University of Toulouse III, Toulouse, France
| | | |
Collapse
|
27
|
Yang W, Deng B, Huang Y, Liu J, Huang Z, Chang Z, Zhu S, Chan LL, Chaudhuri KR, Tan EK, Wang Q. Two heterozygous progranulin mutations in progressive supranuclear palsy. Brain 2021; 144:e27. [PMID: 33844830 DOI: 10.1093/brain/awaa428] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Affiliation(s)
- Wanlin Yang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P. R. China
| | - Bin Deng
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P. R. China
| | - Yu Huang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P. R. China
| | - Jingxing Liu
- Guangzhou Kingmed Center for Clinical Laboratory Co., Ltd, Guangzhou, Guangdong 510005, P. R. China
| | - Zifeng Huang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P. R. China
| | - Zihan Chang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P. R. China
| | - Shuzhen Zhu
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P. R. China
| | - Ling-Ling Chan
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore.,Duke-NUS Medical School, Singapore
| | - K Ray Chaudhuri
- Parkinson Foundation International Centre of Excellence at King's College Hospital, and Kings College, Denmark Hill, London, SE5 9RS UK
| | - Eng-King Tan
- Department of Neurology, National Neuroscience Institute, Singapore General Hospital, Singapore.,Duke-NUS Medical School, Singapore
| | - Qing Wang
- Department of Neurology, Zhujiang Hospital of Southern Medical University, Guangzhou, Guangdong 510282, P. R. China
| |
Collapse
|
28
|
Du H, Wong MY, Zhang T, Santos MN, Hsu C, Zhang J, Yu H, Luo W, Hu F. A multifaceted role of progranulin in regulating amyloid-beta dynamics and responses. Life Sci Alliance 2021; 4:e202000874. [PMID: 34103390 PMCID: PMC8200295 DOI: 10.26508/lsa.202000874] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2020] [Revised: 05/26/2021] [Accepted: 05/26/2021] [Indexed: 01/07/2023] Open
Abstract
Haploinsufficiency of progranulin (PGRN) is a leading cause of frontotemporal lobar degeneration (FTLD). PGRN polymorphisms are associated with Alzheimer's disease. PGRN is highly expressed in the microglia near Aβ plaques and influences plaque dynamics and microglial activation. However, the detailed mechanisms remain elusive. Here we report that PGRN deficiency reduces human APP and Aβ levels in the young male but not female mice. PGRN-deficient microglia exhibit increased expression of markers associated with microglial activation, including CD68, galectin-3, TREM2, and GPNMB, specifically near Aβ plaques. In addition, PGRN loss leads to up-regulation of lysosome proteins and an increase in the nuclear localization of TFE3, a transcription factor involved in lysosome biogenesis. Cultured PGRN-deficient microglia show enhanced nuclear translocation of TFE3 and inflammation in response to Aβ fibril treatment. Taken together, our data revealed a sex- and age-dependent effect of PGRN on APP metabolism and a role of PGRN in regulating lysosomal activities and inflammation in plaque-associated microglia.
Collapse
Affiliation(s)
- Huan Du
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Man Ying Wong
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Tingting Zhang
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Mariela Nunez Santos
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Charlene Hsu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| | - Junke Zhang
- Department of Computational Biology, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Haiyuan Yu
- Department of Computational Biology, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Wenjie Luo
- Feil Family Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, USA
| | - Fenghua Hu
- Department of Molecular Biology and Genetics, Cornell University, Ithaca, NY, USA
| |
Collapse
|
29
|
Kuroda M, Matsuwaki T, Tanaka Y, Yamanouchi K, Nishihara M. Convulsive responses to seizure-inducible drugs are exacerbated in progranulin-deficient mice. Neuroreport 2021; 31:478-483. [PMID: 32168097 DOI: 10.1097/wnr.0000000000001425] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
Progranulin (PGRN) is a glycoprotein that is widely expressed among organs, including the central nervous system. PGRN insufficiency is involved in various neurodegenerative disorders such as frontotemporal dementia, Alzheimer's disease, and neuronal ceroid lipofuscinosis. One of the major causes of neuronal damage is hyperactivation of the cerebrum triggered by upregulation of excitatory systems. In the present study, we examined the possible involvement of PGRN in modulating excitability of the cerebrum using wild type and PGRN-deficient mice. First, we treated wild type and PGRN-deficient mice with seizure-inducible drugs, bicuculline or N-methyl-D-aspartate (NMDA), which provoke hyperexcitement of neurons. PGRN-deficient mice showed higher intensity of seizure and longer duration of convulsive behavior when treated with either bicuculline or NMDA. Next, we quantified the expression of NMDA receptor subunits in the hippocampus and cerebral cortex. The expression level of NR2A subunit protein was significantly higher in the hippocampus of PGRN-deficient mice, while no difference was observed in the cerebral cortex. On the other hand, mRNA levels of NMDA receptor subunits in the hippocampus were comparable or even lower in PGRN-deficient mice. These results suggest that PGRN modulates the excitability of the cerebrum by regulating at least partially the protein level of NMDA receptors in the hippocampus.
Collapse
Affiliation(s)
- Machi Kuroda
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo
| | - Takashi Matsuwaki
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo
| | - Yoshinori Tanaka
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo.,Biochemistry Unit, Faculty of Veterinary Medicine, Okayama University of Science, Ehime, Japan
| | - Keitaro Yamanouchi
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo
| | - Masugi Nishihara
- Department of Veterinary Physiology, Graduate School of Agricultural and Life Sciences, The University of Tokyo, Tokyo
| |
Collapse
|
30
|
Poddar MK, Banerjee S, Chakraborty A, Dutta D. Metabolic disorder in Alzheimer's disease. Metab Brain Dis 2021; 36:781-813. [PMID: 33638805 DOI: 10.1007/s11011-021-00673-z] [Citation(s) in RCA: 19] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/26/2020] [Accepted: 01/14/2021] [Indexed: 12/21/2022]
Abstract
Alzheimer's disease (AD), a well known aging-induced neurodegenerative disease is related to amyloid proteinopathy. This proteinopathy occurs due to abnormalities in protein folding, structure and thereby its function in cells. The root cause of such kind of proteinopathy and its related neurodegeneration is a disorder in metabolism, rather metabolomics of the major as well as minor nutrients. Metabolomics is the most relevant "omics" platform that offers a great potential for the diagnosis and prognosis of neurodegenerative diseases as an individual's metabolome. In recent years, the research on such kinds of neurodegenerative diseases, especially aging-related disorders is broadened its scope towards metabolic function. Different neurotransmitter metabolisms are also involved with AD and its associated neurodegeneration. The genetic and epigenetic backgrounds are also noteworthy. In this review, the physiological changes of AD in relation to its corresponding biochemical, genetic and epigenetic involvements including its (AD) therapeutic aspects are discussed.
Collapse
Affiliation(s)
- Mrinal K Poddar
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, 700032, India.
| | - Soumyabrata Banerjee
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, 700032, India
- Departrment of Psychology, Neuroscience Program, Field Neurosciences Institute Research Laboratory for Restorative Neurology, Central Michigan University, Mount Pleasant, MI, 48859, USA
| | - Apala Chakraborty
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, 700032, India
| | - Debasmita Dutta
- Department of Pharmaceutical Technology, Jadavpur University, 188, Raja S. C. Mallick Road, Kolkata, 700032, India
- Department of Coatings and Polymeric Materials, North Dakota State University, Fargo, ND, 58102, USA
| |
Collapse
|
31
|
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
|
32
|
Boeve BF, Rosen H. Clinical and Neuroimaging Aspects of Familial Frontotemporal Lobar Degeneration Associated with MAPT and GRN Mutations. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2021; 1281:77-92. [PMID: 33433870 DOI: 10.1007/978-3-030-51140-1_6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 04/03/2023]
Abstract
Numerous kindreds with familial frontotemporal lobar degeneration have been linked to mutations in microtubule-associated protein tau (MAPT) or progranulin (GRN) genes. While there are many similarities in the clinical manifestations and associated neuroimaging findings, there are also distinct differences. In this review, we compare and contrast the demographic/inheritance characteristics, histopathology, pathophysiology, clinical aspects, and key neuroimaging findings between those with MAPT and GRN mutations.
Collapse
Affiliation(s)
| | - Howard Rosen
- Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| |
Collapse
|
33
|
Owens LV, Benedetto A, Dawson N, Gaffney CJ, Parkin ET. Gene therapy-mediated enhancement of protective protein expression for the treatment of Alzheimer's disease. Brain Res 2021; 1753:147264. [PMID: 33422539 DOI: 10.1016/j.brainres.2020.147264] [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: 09/24/2020] [Revised: 11/22/2020] [Accepted: 12/20/2020] [Indexed: 12/30/2022]
Abstract
Alzheimer's disease (AD) is the leading form of dementia but lacks curative treatments. Current understanding of AD aetiology attributes the development of the disease to the misfolding of two proteins; amyloid-β (Aβ) and hyperphosphorylated tau, with their pathological accumulation leading to concomitant oxidative stress, neuroinflammation, and neuronal death. These processes are regulated at multiple levels to maintain homeostasis and avert disease. However, many of the relevant regulatory proteins appear to be downregulated in the AD-afflicted brain. Enhancement/restoration of these 'protective' proteins, therefore, represents an attractive therapeutic avenue. Gene therapy is a desirable means of achieving this because it is not associated with the side-effects linked to systemic protein administration, and sustained protein expression virtually eliminates compliance issues. The current article represents a focused and succinct review of the better established 'protective' protein targets for gene therapy enhancement/restoration rather than being designed as an exhaustive review incorporating less validated protein subjects. In addition, we will discuss how the risks associated with uncontrolled or irreversible gene expression might be mitigated through combining neuronal-specific promoters, inducible expression systems and localised injections. Whilst many of the gene therapy targets reviewed herein are yet to enter clinical trials, preclinical testing has thus far demonstrated encouraging potential for the gene therapy-based treatment of AD.
Collapse
Affiliation(s)
- Lauren V Owens
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK
| | - Alexandre Benedetto
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK
| | - Neil Dawson
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK
| | - Christopher J Gaffney
- Lancaster Medical School, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK
| | - Edward T Parkin
- Division of Biomedical and Life Sciences, Faculty of Health and Medicine, Lancaster University, Lancaster LA1 4YG, UK.
| |
Collapse
|
34
|
Bartoletti-Stella A, De Pasqua S, Baiardi S, Bartolomei I, Mengozzi G, Orio G, Pastorelli F, Piras S, Poda R, Raggi A, Stanzani Maserati M, Tarozzi M, Liguori R, Salvi F, Parchi P, Capellari S. Characterization of novel progranulin gene variants in Italian patients with neurodegenerative diseases. Neurobiol Aging 2021; 97:145.e7-145.e15. [PMID: 32507413 DOI: 10.1016/j.neurobiolaging.2020.05.004] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 04/19/2020] [Accepted: 05/05/2020] [Indexed: 12/14/2022]
Abstract
Loss-of-function mutations in the gene encoding for the protein progranulin (PGRN), GRN, are one of the major genetic abnormalities involved in frontotemporal lobar degeneration. However, genetic variations, mainly missense, in GRN have also been linked to other neurodegenerative diseases. We found 12 different pathogenic/likely pathogenic variants in 21 patients identified in a cohort of Italian patients affected by various neurodegenerative disorders. We detected the p.Thr272SerfsTer10 as the most frequent, followed by the c.1179+3A>G variant. We characterized the clinical phenotype of 12 patients from 3 pedigrees carrying the c.1179+3A>G variant, demonstrated the pathogenicity of this mutation, and detected other rarer variants causing haploinsufficiency (p.Met1?, c.709-2A>T, p.Gly79AspfsTer39). Finally, by applying bioinformatics, neuropathological, and biochemical studies, we characterized 6 missense/synonymous variants (p.Asp94His, p.Gly117Asp, p.Ala266Pro, p.Val279Val, p.Arg298His, p.Ala505Gly), including 4 previously unreported. The designation of variants is crucial for genetic counseling and the enrollment of patients in clinical studies.
Collapse
Affiliation(s)
| | - Silvia De Pasqua
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
| | - Simone Baiardi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy; Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Ilaria Bartolomei
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
| | - Giacomo Mengozzi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
| | - Giuseppe Orio
- Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Francesca Pastorelli
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
| | - Silvia Piras
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
| | - Roberto Poda
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
| | - Alberto Raggi
- Unità Operativa di Neurologia, Ospedale G.B. Morgagni - L. Pierantoni, Forlì, Italy
| | | | - Martina Tarozzi
- Department of Medical and Surgical Sciences (DIMEC), University of Bologna, Bologna, Italy
| | - Rocco Liguori
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy; Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy
| | - Fabrizio Salvi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy
| | - Piero Parchi
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy; Department of Diagnostic Experimental and Specialty Medicine (DIMES), University of Bologna, Bologna, Italy
| | - Sabina Capellari
- IRCCS Istituto delle Scienze Neurologiche di Bologna, Bellaria Hospital, Bologna, Italy; Department of Biomedical and NeuroMotor Sciences (DIBINEM), University of Bologna, Bologna, Italy.
| |
Collapse
|
35
|
Calabrò M, Rinaldi C, Santoro G, Crisafulli C. The biological pathways of Alzheimer disease: a review. AIMS Neurosci 2020; 8:86-132. [PMID: 33490374 PMCID: PMC7815481 DOI: 10.3934/neuroscience.2021005] [Citation(s) in RCA: 127] [Impact Index Per Article: 25.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2020] [Accepted: 11/23/2020] [Indexed: 12/18/2022] Open
Abstract
Alzheimer disease is a progressive neurodegenerative disorder, mainly affecting older people, which severely impairs patients' quality of life. In the recent years, the number of affected individuals has seen a rapid increase. It is estimated that up to 107 million subjects will be affected by 2050 worldwide. Research in this area has revealed a lot about the biological and environmental underpinnings of Alzheimer, especially its correlation with β-Amyloid and Tau related mechanics; however, the precise molecular events and biological pathways behind the disease are yet to be discovered. In this review, we focus our attention on the biological mechanics that may lie behind Alzheimer development. In particular, we briefly describe the genetic elements and discuss about specific biological processes potentially associated with the disease.
Collapse
Affiliation(s)
| | | | | | - Concetta Crisafulli
- Department of Biomedical and Dental Sciences and Morphofunctional Imaging, University of Messina, Italy
| |
Collapse
|
36
|
Tantawy AA, Adly AA, Ismail EA, Salah NY, Abdel Alem S, Elsantiel HI. Serum progranulin levels in paediatric patients with Gaucher disease; relation to disease severity and liver stiffness by transient elastography. Liver Int 2020; 40:3051-3060. [PMID: 32652633 DOI: 10.1111/liv.14598] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/28/2019] [Revised: 06/15/2020] [Accepted: 07/01/2020] [Indexed: 12/20/2022]
Abstract
BACKGROUND Non-invasive screening for liver fibrosis using transient elastography (TE) could be of value in the management of Gaucher disease (GD). Progranulin (PGRN) is a novel disease modifier in GD and an independent marker of liver fibrosis. OBJECTIVES We determined PGRN levels in paediatric patients with GD and assessed its role as a potential marker for disease severity and relation to liver stiffness by TE. METHODS Fifty-one GD patients (20 had type 1 and 31 had type 3) with a median age of 9.5 years were compared to 40 age- and sex-matched healthy controls and were studied focusing on visceral manifestations, neurological disease, haematological profile and PGRN levels as well as abdominal ultrasound and TE. Patients were on enzyme replacement therapy (ERT) for various durations and those with viral hepatitis infection were excluded. RESULTS By TE, 14 GD patients (27.5%) had elevated liver stiffness ≥7.0 kPa. Liver stiffness was significantly higher in type 1 GD patients than type 3 (P = .002), in splenectomized patients (P = .012) and those with dysphagia (P < .001). Liver stiffness was positively correlated with age of onset of ERT (P < .001). PGRN levels were significantly lower in GD patients compared with controls (P < .001). PGRN was significantly lower in GD patients with squint (P = .025), dysphagia (P = .036) and elevated liver stiffness (P = .015). PGRN was positively correlated with white blood cell count (r = .455, P = .002) and haemoglobin (r = .546, P < .001), while negatively correlated with severity score index (r = -.529, P < .001), liver volume (r = -.298, P = .034) and liver stiffness (r = -.652, P < .001). CONCLUSIONS Serum PGRN levels were associated with clinical disease severity and elevated liver stiffness in paediatric GD patients.
Collapse
Affiliation(s)
- Azza A Tantawy
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Amira A Adly
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Eman A Ismail
- Clinical Pathology Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Nouran Y Salah
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| | - Shereen Abdel Alem
- Department of Endemic medicine and Hepatology, Faculty of medicine, Cairo University, Cairo, Egypt
| | - Hesham I Elsantiel
- Pediatrics Department, Faculty of Medicine, Ain Shams University, Cairo, Egypt
| |
Collapse
|
37
|
Giunta M, Libri I, Premi E, Brattini C, Paghera B, Archetti S, Gasparotti R, Padovani A, Borroni B, Benussi A. Clinical and radiological features of posterior cortical atrophy (PCA) in a GRN mutation carrier: a case report. Eur J Neurol 2020; 28:344-348. [PMID: 33030763 DOI: 10.1111/ene.14574] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2020] [Accepted: 10/01/2020] [Indexed: 11/28/2022]
Abstract
BACKGROUND AND PURPOSE Posterior cortical atrophy (PCA) is a rare neurodegenerative syndrome, defined by a distinctive clinical-radiological profile, with Alzheimer's disease (AD) pathology accounting for the majority of cases. The aim of this report was to present the case of a patient with impairment of visual and constructional abilities as initial manifestations. METHOD The patient underwent a multidimensional assessment, including neuropsychological evaluation, structural and functional imaging and genetic screening. RESULTS Neurological and neuropsychological assessment showed an impairment of constructive and visuo-spatial skills, associated with dyscalculia, simultanagnosia, optic ataxia and oculomotor apraxia. In accordance with the latest consensus criteria, a diagnosis of PCA was made. Consistent with the clinical findings, structural and functional imaging showed a peculiar pattern of atrophy with primary involvement of right parieto-occipital cortices, whereas cerebrospinal fluid biochemical analysis did not reveal a profile compatible with AD pathology. Genetic screening identified a known pathogenic GRN mutation. CONCLUSION We present a case of PCA in a GRN mutation carrier in whom a concomitant AD pathological process was excluded. Consequently, although lacking histological data, our case suggests GRN-related pathology causative of PCA. Through this report we provide further evidence for a new neurodegenerative pathway leading to PCA, extending the clinical spectrum of GRN-associated phenotypes.
Collapse
Affiliation(s)
- M Giunta
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - I Libri
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - E Premi
- Stroke Unit, ASST Spedali Civili, Brescia, Italy
| | - C Brattini
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - B Paghera
- Nuclear Medicine Department, University of Brescia, Brescia, Italy
| | - S Archetti
- Biotechnology Laboratory, Department of Diagnostics, Spedali Civili Hospital, Brescia, Italy
| | - R Gasparotti
- Neuroradiology Unit, University of Brescia, Brescia, Italy
| | - A Padovani
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - B Borroni
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| | - A Benussi
- Centre for Neurodegenerative Disorders, Department of Clinical and Experimental Sciences, University of Brescia, Brescia, Italy
| |
Collapse
|
38
|
Jiang S, Bhaskar K. Degradation and Transmission of Tau by Autophagic-Endolysosomal Networks and Potential Therapeutic Targets for Tauopathy. Front Mol Neurosci 2020; 13:586731. [PMID: 33177989 PMCID: PMC7596180 DOI: 10.3389/fnmol.2020.586731] [Citation(s) in RCA: 65] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2020] [Accepted: 09/24/2020] [Indexed: 01/21/2023] Open
Abstract
Tauopathies are a class of neurodegenerative diseases, including Alzheimer’s disease (AD), Frontotemporal Dementia (FTD), Progressive Supranuclear Palsy (PSP), Corticobasal Degeneration (CBD), and many others where microtubule-associated protein tau (MAPT or tau) is hyperphosphorylated and aggregated to form insoluble paired helical filaments (PHFs) and ultimately neurofibrillary tangles (NFTs). Autophagic-endolysosomal networks (AELN) play important roles in tau clearance. Excessive soluble neurotoxic forms of tau and tau hyperphosphorylated at specific sites are cleared through the ubiquitin-proteasome system (UPS), Chaperon-mediated Autophagy (CMA), and endosomal microautophagy (e-MI). On the other hand, intra-neuronal insoluble tau aggregates are often degraded within lysosomes by macroautophagy. AELN defects have been observed in AD, FTD, CBD, and PSP, and lysosomal dysfunction was shown to promote the cleavage and neurotoxicity of tau. Moreover, several AD risk genes (e.g., PICALM, GRN, and BIN1) have been associated with dysregulation of AELN in the late-onset sporadic AD. Conversely, tau dissociation from microtubules interferes with retrograde transport of autophagosomes to lysosomes, and that tau fragments can also lead to lysosomal dysfunction. Recent studies suggest that tau is not merely an intra-neuronal protein, but it can be released to brain parenchyma via extracellular vesicles, like exosomes and ectosomes, and thus spread between neurons. Extracellular tau can also be taken up by microglial cells and astrocytes, either being degraded through AELN or propagated via exosomes. This article reviews the complex roles of AELN in the degradation and transmission of tau, potential diagnostic/therapeutic targets and strategies based on AELN-mediated tau clearance and propagation, and the current state of drug development targeting AELN and tau against tauopathies.
Collapse
Affiliation(s)
- Shanya Jiang
- Department of Molecular Genetics and Microbiology, The University of New Mexico, Albuquerque, NM, United States
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, The University of New Mexico, Albuquerque, NM, United States
| |
Collapse
|
39
|
Nguyen AD, Malmstrom TK, Niehoff ML, Aziz A, Miller DK, Morley JE. Serum progranulin levels are associated with frailty in middle-aged individuals. PLoS One 2020; 15:e0238877. [PMID: 32886731 PMCID: PMC7473561 DOI: 10.1371/journal.pone.0238877] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2020] [Accepted: 08/25/2020] [Indexed: 01/08/2023] Open
Abstract
BACKGROUND AND OBJECTIVE A recent study identified progranulin as a candidate biomarker for frailty, based on gene expression databases. In the present study, we investigated associations between serum progranulin levels and frailty in a population-based sample of late middle-age and older adults. METHODS We utilized a cohort study that included 358 African Americans (baseline ages 49-65). Frailty was assessed by three established methods: the interview-based FRAIL scale, the Cardiovascular Health Study (CHS) frailty scale that includes performance-based measurements, and the Frailty Index (FI) that is based on cumulative deficits. Serum levels of the following proteins and metabolites were measured: progranulin, cystatin C, fructosamine, soluble cytokine receptors (interleukin-2 and -6, tumor necrosis factor α-1 and -2), and C-reactive protein. Sarcopenia was assessed using the SARC-F index. Vital status was determined by matching through the National Death Index (NDI). RESULTS Serum progranulin levels were associated with frailty for all indices (FRAIL, CHS, and FI) but not with sarcopenia. Inflammatory markers indicated by soluble cytokine receptors (sIL-2R, sIL-6R, sTNFR1, sTNFR2) were positively associated serum progranulin. Increased serum progranulin levels at baseline predicted poorer outcomes including future frailty as measured by the FRAIL scale and 15-year all-cause mortality independent of age, gender, and frailty. CONCLUSIONS Our findings suggest that serum progranulin levels may be a candidate biomarker for physical frailty, independent of sarcopenia. Further studies are needed to validate this association and assess the utility of serum progranulin levels as a potential biomarker for prevalent frailty, for risk for developing incident frailty, and for mortality risk over and above the effect of baseline frailty.
Collapse
Affiliation(s)
- 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
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, Missouri, United States of America
| | - Theodore K. Malmstrom
- Henry and Amelia Nasrallah Center for Neuroscience, Saint Louis University, St. Louis, Missouri, United States of America
- Department of Psychiatry and Behavioral Neuroscience, Saint Louis University School of Medicine, 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
| | - Asef Aziz
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| | - Douglas K. Miller
- Regenstrief Institute, Inc., Indianapolis, Indiana, United States of America
| | - John E. Morley
- Division of Geriatric Medicine, Department of Internal Medicine, Saint Louis University School of Medicine, St. Louis, Missouri, United States of America
| |
Collapse
|
40
|
Häkkinen S, Chu SA, Lee SE. Neuroimaging in genetic frontotemporal dementia and amyotrophic lateral sclerosis. Neurobiol Dis 2020; 145:105063. [PMID: 32890771 DOI: 10.1016/j.nbd.2020.105063] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Revised: 07/30/2020] [Accepted: 08/26/2020] [Indexed: 02/06/2023] Open
Abstract
Frontotemporal dementia (FTD) and amyotrophic lateral sclerosis (ALS) have a strong clinical, genetic and pathological overlap. This review focuses on the current understanding of structural, functional and molecular neuroimaging signatures of genetic FTD and ALS. We overview quantitative neuroimaging studies on the most common genes associated with FTD (MAPT, GRN), ALS (SOD1), and both (C9orf72), and summarize visual observations of images reported in the rarer genes (CHMP2B, TARDBP, FUS, OPTN, VCP, UBQLN2, SQSTM1, TREM2, CHCHD10, TBK1).
Collapse
Affiliation(s)
- Suvi Häkkinen
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Stephanie A Chu
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA
| | - Suzee E Lee
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, San Francisco, CA, USA.
| |
Collapse
|
41
|
Guan Z, Chen Z, Fu S, Dai L, Shen Y. Progranulin Administration Attenuates β-Amyloid Deposition in the Hippocampus of 5xFAD Mice Through Modulating BACE1 Expression and Microglial Phagocytosis. Front Cell Neurosci 2020; 14:260. [PMID: 32973454 PMCID: PMC7461932 DOI: 10.3389/fncel.2020.00260] [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: 03/01/2020] [Accepted: 07/27/2020] [Indexed: 01/08/2023] Open
Abstract
Loss of function mutations in the progranulin (PGRN) gene is a risk factor for Alzheimer’s disease (AD). Previous works reported that the deficiency of PGRN accelerates β-amyloid (Aβ) accumulation in AD transgenic mouse brains while overexpression of PGRN could restrain disease progression. However, mechanisms of PGRN in protecting against Aβ deposition remains unclear. Here, using the 5xFAD AD mouse model, we show that intrahippocampal injection of PGRN protein leads to a reduction of Aβ plaques, downregulation of beta-secretase 1 (BACE1), and enhanced microglia Aβ phagocytosis in the mouse hippocampus. Furthermore, PGRN treatment inhibited BACE1 expression in N2a cells and primary culture neurons and improved the phagocytic capacity of microglia isolated from 5xFAD mouse brains. Collectively, our results provide further evidence that enhancing progranulin could be a promising option for AD therapy.
Collapse
Affiliation(s)
- Zhangxin Guan
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Zuolong Chen
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Shumei Fu
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Linbin Dai
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, School of Life Sciences, University of Science and Technology of China, Hefei, China
| | - Yong Shen
- Institute on Aging and Brain Disorders, The First Affiliated Hospital of USTC, Division of Life Sciences and Medicine, University of Science and Technology of China, Hefei, China.,Hefei National Laboratory for Physical Sciences at the Microscale, Neurodegenerative Disorder Research Center, School of Life Sciences, University of Science and Technology of China, Hefei, China.,Center for Excellence in Brain Science and Intelligence Technology, Chinese Academy of Sciences, Shanghai, China
| |
Collapse
|
42
|
Huin V, Barbier M, Bottani A, Lobrinus JA, Clot F, Lamari F, Chat L, Rucheton B, Fluchère F, Auvin S, Myers P, Gelot A, Camuzat A, Caillaud C, Jornéa L, Forlani S, Saracino D, Duyckaerts C, Brice A, Durr A, Le Ber I. Homozygous GRN mutations: new phenotypes and new insights into pathological and molecular mechanisms. Brain 2020; 143:303-319. [PMID: 31855245 DOI: 10.1093/brain/awz377] [Citation(s) in RCA: 46] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Revised: 10/08/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
Homozygous mutations in the progranulin gene (GRN) are associated with neuronal ceroid lipofuscinosis 11 (CLN11), a rare lysosomal-storage disorder characterized by cerebellar ataxia, seizures, retinitis pigmentosa, and cognitive disorders, usually beginning between 13 and 25 years of age. This is a rare condition, previously reported in only four families. In contrast, heterozygous GRN mutations are a major cause of frontotemporal dementia associated with neuronal cytoplasmic TDP-43 inclusions. We identified homozygous GRN mutations in six new patients. The phenotypic spectrum is much broader than previously reported, with two remarkably distinct presentations, depending on the age of onset. A childhood/juvenile form is characterized by classical CLN11 symptoms at an early age at onset. Unexpectedly, other homozygous patients presented a distinct delayed phenotype of frontotemporal dementia and parkinsonism after 50 years; none had epilepsy or cerebellar ataxia. Another major finding of this study is that all GRN mutations may not have the same impact on progranulin protein synthesis. A hypomorphic effect of some mutations is supported by the presence of residual levels of plasma progranulin and low levels of normal transcript detected in one case with a homozygous splice-site mutation and late onset frontotemporal dementia. This is a new critical finding that must be considered in therapeutic trials based on replacement strategies. The first neuropathological study in a homozygous carrier provides new insights into the pathological mechanisms of the disease. Hallmarks of neuronal ceroid lipofuscinosis were present. The absence of TDP-43 cytoplasmic inclusions markedly differs from observations of heterozygous mutations, suggesting a pathological shift between lysosomal and TDP-43 pathologies depending on the mono or bi-allelic status. An intriguing observation was the loss of normal TDP-43 staining in the nucleus of some neurons, which could be the first stage of the TDP-43 pathological process preceding the formation of typical cytoplasmic inclusions. Finally, this study has important implications for genetic counselling and molecular diagnosis. Semi-dominant inheritance of GRN mutations implies that specific genetic counselling should be delivered to children and parents of CLN11 patients, as they are heterozygous carriers with a high risk of developing dementia. More broadly, this study illustrates the fact that genetic variants can lead to different phenotypes according to their mono- or bi-allelic state, which is a challenge for genetic diagnosis.
Collapse
Affiliation(s)
- Vincent Huin
- Sorbonne Université, Institut du Cerveau et de la Moelle épinière (ICM), AP-HP, INSERM, CNRS, University Hospital Pitié-Salpêtrière, Paris, France
| | - Mathieu Barbier
- Sorbonne Université, Institut du Cerveau et de la Moelle épinière (ICM), AP-HP, INSERM, CNRS, University Hospital Pitié-Salpêtrière, Paris, France
| | - Armand Bottani
- Department of Genetic Medicine, University Hospital of Geneva, Geneva, Switzerland
| | | | - Fabienne Clot
- Department of Molecular and Cellular Neurogenetics, AP-HP, Pitié-Salpêtrière - Charles Foix University Hospitals, Paris, France
| | - Foudil Lamari
- AP-HP, Metabolic Biochemistry Unit, Department of Biochemistry of Neurometabolic Diseases, Pitié-Salpêtrière University Hospital, Paris, France
| | - Laureen Chat
- Department of Molecular and Cellular Neurogenetics, AP-HP, Pitié-Salpêtrière - Charles Foix University Hospitals, Paris, France
| | - Benoît Rucheton
- AP-HP, Metabolic Biochemistry Unit, Department of Biochemistry of Neurometabolic Diseases, Pitié-Salpêtrière University Hospital, Paris, France
| | - Frédérique Fluchère
- AP-HM, Department of Neurology and Movement Disorders, La Timone, Clinical Neuroscience Unit, Aix-Marseille University, France
| | - Stéphane Auvin
- AP-HP Department of Pediatric Neurology, Robert Debré University Hospital, F, Paris, France
| | | | - Antoinette Gelot
- Neuropathology, Department of Pathology, Trusseau Hospital, AP-HP, Paris, France and INMED INSERM U901 Luminy Campus, Aix-Marseille University, France
| | - Agnès Camuzat
- Sorbonne Université, Institut du Cerveau et de la Moelle épinière (ICM), AP-HP, INSERM, CNRS, University Hospital Pitié-Salpêtrière, Paris, France
| | - Catherine Caillaud
- Biochemical, Metabolomical and Proteonomical Department, Necker-Enfants Malades University Hospital, AP-HP, F-75015 Paris, France
| | - Ludmila Jornéa
- Sorbonne Université, Institut du Cerveau et de la Moelle épinière (ICM), AP-HP, INSERM, CNRS, University Hospital Pitié-Salpêtrière, Paris, France
| | - Sylvie Forlani
- Sorbonne Université, Institut du Cerveau et de la Moelle épinière (ICM), AP-HP, INSERM, CNRS, University Hospital Pitié-Salpêtrière, Paris, France
| | - Dario Saracino
- Sorbonne Université, Institut du Cerveau et de la Moelle épinière (ICM), AP-HP, INSERM, CNRS, University Hospital Pitié-Salpêtrière, Paris, France
| | - Charles Duyckaerts
- Department of Neuropathology 'Escourolle', AP-HP, Pitié-Salpêtrière University Hospital, Paris, France
| | - Alexis Brice
- Sorbonne Université, Institut du Cerveau et de la Moelle épinière (ICM), AP-HP, INSERM, CNRS, University Hospital Pitié-Salpêtrière, Paris, France.,AP-HP, National Reference Center for Rare Diseases 'Neurogenetics', Pitié-Salpêtrière University Hospital, Paris, France
| | - Alexandra Durr
- Sorbonne Université, Institut du Cerveau et de la Moelle épinière (ICM), AP-HP, INSERM, CNRS, University Hospital Pitié-Salpêtrière, Paris, France.,AP-HP, National Reference Center for Rare Diseases 'Neurogenetics', Pitié-Salpêtrière University Hospital, Paris, France
| | - Isabelle Le Ber
- Sorbonne Université, Institut du Cerveau et de la Moelle épinière (ICM), AP-HP, INSERM, CNRS, University Hospital Pitié-Salpêtrière, Paris, France.,AP-HP, National Reference center 'rare and young dementias', IM2A, Pitié-Salpêtrière University Hospital, Paris, France
| |
Collapse
|
43
|
Feng T, Mai S, Roscoe JM, Sheng RR, Ullah M, Zhang J, Katz II, Yu H, Xiong W, Hu F. Loss of TMEM106B and PGRN leads to severe lysosomal abnormalities and neurodegeneration in mice. EMBO Rep 2020; 21:e50219. [PMID: 32852886 PMCID: PMC7534636 DOI: 10.15252/embr.202050219] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2020] [Revised: 06/25/2020] [Accepted: 07/14/2020] [Indexed: 12/12/2022] Open
Abstract
Haploinsufficiency of progranulin (PGRN) is a leading cause of frontotemporal lobar degeneration (FTLD). Loss of PGRN leads to lysosome dysfunction during aging. TMEM106B, a gene encoding a lysosomal membrane protein, is the main risk factor for FTLD with PGRN haploinsufficiency. But how TMEM106B affects FTLD disease progression remains to be determined. Here, we report that TMEM106B deficiency in mice leads to accumulation of lysosome vacuoles at the distal end of the axon initial segment in motor neurons and the development of FTLD‐related pathology during aging. Ablation of both PGRN and TMEM106B in mice results in severe neuronal loss and glial activation in the spinal cord, retina, and brain. Enlarged lysosomes are frequently found in both microglia and astrocytes. Loss of both PGRN and TMEM106B results in an increased accumulation of lysosomal vacuoles in the axon initial segment of motor neurons and enhances the manifestation of FTLD phenotypes with a much earlier onset. These results provide novel insights into the role of TMEM106B in the lysosome, in brain aging, and in FTLD pathogenesis.
Collapse
Affiliation(s)
- Tuancheng Feng
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Shuyi Mai
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Jenn Marie Roscoe
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Rory R Sheng
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Mohammed Ullah
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Junke Zhang
- Department of Computational Biology, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Isabel Iscol Katz
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Haiyuan Yu
- Department of Computational Biology, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| | - Wenjun Xiong
- Department of Biomedical Sciences, City University of Hong Kong, Kowloon, Hong Kong SAR, China
| | - Fenghua Hu
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY, USA
| |
Collapse
|
44
|
Guven G, Bilgic B, Tufekcioglu Z, Erginel Unaltuna N, Hanagasi H, Gurvit H, Singleton A, Hardy J, Emre M, Gulec C, Bras J, Guerreiro R, Lohmann E. Peripheral GRN mRNA and Serum Progranulin Levels as a Potential Indicator for Both the Presence of Splice Site Mutations and Individuals at Risk for Frontotemporal Dementia. J Alzheimers Dis 2020; 67:159-167. [PMID: 30475763 DOI: 10.3233/jad-180599] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Progranulin (GRN) gene mutations are a major cause of frontotemporal dementia (FTD). Most mutations identified to date are null mutations, which are predicted to cause the pathology via haploinsufficiency. Decreased peripheral progranulin protein (PGRN) levels are associated with the presence of GRN null mutations and are accepted as reliable biomarkers. In this study, our aim was to test whether the presence of specific GRN splice site mutations (c.- 8+2T>G and c.708+6_9del), could be predicted by peripheral mRNA or protein GRN levels, by studying affected and asymptomatic individuals from FTD families. We also tested four missense GRN variants to assess if altered GRN levels depended on the type of mutation.Our results confirmed a reduction in both mRNA and protein PGRN levels in the splice site mutation carriers, which is consistent with previous reports for null mutations. Our results also suggested that both decreased peripheral GRN mRNA and serum PGRN levels indicate the presence of pathogenic mutations in affected individuals, and identify the asymptomatic individuals at risk, without previous knowledge of genetic status. Both inferences suggest a potential use of peripheral GRN mRNA or serum PGRN levels as biomarkers for families with FTD.
Collapse
Affiliation(s)
- Gamze Guven
- Department of Genetics, Aziz Sancar Institute for Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Başar Bilgic
- Department of Neurology, Behavioural Neurology and Movement Disorders Unit, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Zeynep Tufekcioglu
- Department of Neurology, Behavioural Neurology and Movement Disorders Unit, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Nihan Erginel Unaltuna
- Department of Genetics, Aziz Sancar Institute for Experimental Medicine, Istanbul University, Istanbul, Turkey
| | - Hasmet Hanagasi
- Department of Neurology, Behavioural Neurology and Movement Disorders Unit, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Hakan Gurvit
- Department of Neurology, Behavioural Neurology and Movement Disorders Unit, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Andrew Singleton
- Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
| | - John Hardy
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK
| | - Murat Emre
- Department of Neurology, Behavioural Neurology and Movement Disorders Unit, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Cagri Gulec
- Department of Medical Genetics, Istanbul Faculty of Medicine, Istanbul University, Istanbul, Turkey
| | - Jose Bras
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK.,Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal.,UK Dementia Research Institute at UCL (UK DRI), London, UK
| | - Rita Guerreiro
- Department of Molecular Neuroscience, Institute of Neurology, University College London, London, UK.,Department of Medical Sciences and Institute of Biomedicine - iBiMED, University of Aveiro, Aveiro, Portugal.,UK Dementia Research Institute at UCL (UK DRI), London, UK
| | - Ebba Lohmann
- Department of Neurodegenerative Diseases, Hertie Institute for Clinical Brain Research, University of Tübingen, Germany.,DZNE, German Center for Neurodegenerative Diseases, Tübingen, Germany
| |
Collapse
|
45
|
Elia LP, Reisine T, Alijagic A, Finkbeiner S. Approaches to develop therapeutics to treat frontotemporal dementia. Neuropharmacology 2020; 166:107948. [PMID: 31962288 DOI: 10.1016/j.neuropharm.2020.107948] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2019] [Revised: 12/16/2019] [Accepted: 01/07/2020] [Indexed: 12/13/2022]
Abstract
Frontotemporal degeneration (FTD) is a complex disease presenting as a spectrum of clinical disorders with progressive degeneration of frontal and temporal brain cortices and extensive neuroinflammation that result in personality and behavior changes, and eventually, death. There are currently no effective therapies for FTD. While 60-70% of FTD patients are sporadic cases, the other 30-40% are heritable (familial) cases linked to mutations in several known genes. We focus here on FTD caused by mutations in the GRN gene, which encodes a secreted protein, progranulin (PGRN), that has diverse roles in regulating cell survival, immune responses, and autophagy and lysosome function in the brain. FTD-linked mutations in GRN reduce brain PGRN levels that lead to autophagy and lysosome dysfunction, TDP43 accumulation, excessive microglial activation, astrogliosis, and neuron death through still poorly understood mechanisms. PGRN insufficiency has also been linked to Alzheimer's disease (AD), and so the development of therapeutics for GRN-linked FTD that restore PGRN levels and function may have broader application for other neurodegenerative diseases. This review focuses on a strategy to increase PGRN to functional, healthy levels in the brain by identifying novel genetic and chemical modulators of neuronal PGRN levels. This article is part of the special issue entitled 'The Quest for Disease-Modifying Therapies for Neurodegenerative Disorders'.
Collapse
Affiliation(s)
- Lisa P Elia
- Center for Systems and Therapeutics and Taube/Koret Center for Neurodegenerative Disease Research, San Francisco, CA, USA; The J. David Gladstone Institutes, San Francisco, CA, USA.
| | - Terry Reisine
- Independent Scientific Consultant, Santa Cruz, CA, USA
| | - Amela Alijagic
- Center for Systems and Therapeutics and Taube/Koret Center for Neurodegenerative Disease Research, San Francisco, CA, USA; The J. David Gladstone Institutes, San Francisco, CA, USA
| | - Steven Finkbeiner
- Center for Systems and Therapeutics and Taube/Koret Center for Neurodegenerative Disease Research, San Francisco, CA, USA; The J. David Gladstone Institutes, San Francisco, CA, USA; Departments of Neurology and Physiology, UCSF, San Francisco, CA, USA.
| |
Collapse
|
46
|
Abstract
PURPOSE OF REVIEW This article describes the clinical, anatomic, genetic, and pathologic features of behavioral variant frontotemporal dementia (bvFTD) and discusses strategies to improve diagnostic accuracy, emphasizing common pitfalls to avoid. Key aspects of management and the future of diagnosis and care for the disorder are highlighted. RECENT FINDINGS BvFTD is a clinical syndrome, not a disease. Patients with the syndrome share core symptoms that reflect degeneration within the most consistently affected brain regions, but accompanying features vary and reflect the precise topography of regional degeneration. The clinician must distinguish a bvFTD syndrome from psychiatric illness and other neurodegenerative syndromes that feature a prominent behavioral component. Antemortem prediction of pathologic diagnosis remains imperfect but improves with careful attention to the clinical details. Management should emphasize prevention of caregiver distress, behavioral and environmental strategies, symptom-based psychopharmacology, and genetic counseling. SUMMARY BvFTD is an important and challenging dementia syndrome. Although disease-modifying treatments are lacking, clinicians can have a profound impact on a family coping with this disorder. Treatment trials are under way for some genetic forms of bvFTD. For sporadic disease, pathologic heterogeneity remains a major challenge, and ongoing research seeks to improve antemortem molecular diagnosis to facilitate therapeutic discovery.
Collapse
|
47
|
Novel GRN mutations in Koreans with Alzheimer’s disease. Mol Cell Toxicol 2019. [DOI: 10.1007/s13273-019-0038-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
|
48
|
Giau VV, Bagyinszky E, Yang YS, Youn YC, An SSA, Kim SY. Genetic analyses of early-onset Alzheimer's disease using next generation sequencing. Sci Rep 2019; 9:8368. [PMID: 31182772 PMCID: PMC6557896 DOI: 10.1038/s41598-019-44848-2] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2018] [Accepted: 05/23/2019] [Indexed: 12/24/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common type of neurodegenerative dementia, but the cause of AD remained poorly understood. Many mutations in the amyloid precursor protein (APP) and presenilin 1 and 2 (PSEN1 and PSEN2) have been reported as the pathogenic causes of early-onset AD (EOAD), which accounts for up to 5% of all AD cases. In this study, we screened familiar/de novo EOAD (n = 67) samples by next-generation sequencing (NGS) of a 50-gene panel, which included causative and possible pathogenic variants linked to neurodegenerative disorders. Remarkably, three missense mutations in PSEN1 (T119I, G209A, and G417A) and one known variant in PSEN2 (H169N) were discovered in 6% of the cases. Additionally, 67 missense mutations in susceptibility genes for late-onset AD were identified, which may be involved in cholesterol transport, inflammatory response, and β-amyloid modulation. We identified 70 additional novel and missense variants in other genes, such as MAPT, GRN, CSF1R, and PRNP, related to neurodegenerative diseases, which may represent overlapping clinical and neuropathological features with AD. Extensive genetic screening of Korean patients with EOAD identified multiple rare variants with potential roles in AD pathogenesis. This study suggests that individuals diagnosed with AD should be screened for other neurodegenerative disease-associated genes. Our findings expand the classic set of genes involved in neurodegenerative pathogenesis, which should be screened for in clinical trials. Main limitation of this study was the absence of functional assessment for possibly and probably pathogenic variants. Additional issues were that we could not perform studies on copy number variants, and we could not verify the segregation of mutations.
Collapse
Affiliation(s)
- Vo Van Giau
- Department of Bionano Technology, Gachon University, Seongnam, 13120, South Korea
| | - Eva Bagyinszky
- Department of Bionano Technology, Gachon University, Seongnam, 13120, South Korea
| | - Young Soon Yang
- Department of Neurology, Veterans Health Service Medical Center, Seoul, 05368, South Korea
| | - Young Chul Youn
- Department of Neurology, Chung-Ang University Hospital, Seoul, 06973, South Korea
| | - Seong Soo A An
- Department of Bionano Technology, Gachon University, Seongnam, 13120, South Korea.
| | - Sang Yun Kim
- Department of Neurology, Seoul National University College of Medicine & Neurocognitive Behavior Center, Seoul National University Bundang Hospital, Seongnam, 13620, South Korea.
| |
Collapse
|
49
|
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
|
50
|
Ramos EM, Dokuru DR, Van Berlo V, Wojta K, Wang Q, Huang AY, Miller ZA, Karydas AM, Bigio EH, Rogalski E, Weintraub S, Rader B, Miller BL, Gorno-Tempini ML, Mesulam MM, Coppola G. Genetic screen in a large series of patients with primary progressive aphasia. Alzheimers Dement 2019; 15:553-560. [PMID: 30599136 PMCID: PMC6480353 DOI: 10.1016/j.jalz.2018.10.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2018] [Revised: 10/05/2018] [Accepted: 10/30/2018] [Indexed: 12/12/2022]
Abstract
INTRODUCTION Primary progressive aphasia (PPA) is a neurological syndrome, associated with both frontotemporal dementia and Alzheimer's disease, in which progressive language impairment emerges as the most salient clinical feature during the initial stages of disease. METHODS We screened the main genes associated with Alzheimer's disease and frontotemporal dementia for pathogenic and risk variants in a cohort of 403 PPA cases. RESULTS In this case series study, 14 (3.5%) cases carried (likely) pathogenic variants: four C9orf72 expansions, nine GRN, and one TARDBP mutation. Rare risk variants, TREM2 R47H and MAPT A152T, were associated with a three- to seven-fold increase in risk for PPA. DISCUSSION Our results show that while pathogenic variants within the most common dementia genes were rarely associated with PPA, these were found almost exclusively in GRN and C9orf72, suggesting that PPA is more TDP43- than tau-related in our series. This is consistent with the finding that PPA frequency in dominantly inherited dementias is the highest in kindreds with GRN variants.
Collapse
Affiliation(s)
- Eliana Marisa Ramos
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Deepika Reddy Dokuru
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Victoria Van Berlo
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Kevin Wojta
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Qing Wang
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Alden Y Huang
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA; Bioinformatics Interdepartmental Program, University of California, Los Angeles, Los Angeles, CA, USA
| | - Zachary A Miller
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Anna M Karydas
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Eileen H Bigio
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Emily Rogalski
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Sandra Weintraub
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Benjamin Rader
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Bruce L Miller
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Maria Luisa Gorno-Tempini
- Memory and Aging Center, Department of Neurology, University of California San Francisco, San Francisco, CA, USA
| | - Marek-Marsel Mesulam
- Mesulam Center for Cognitive Neurology and Alzheimer's Disease, Northwestern University, Chicago, IL, USA
| | - Giovanni Coppola
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA.
| |
Collapse
|