1
|
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
|
2
|
Colella P, Sayana R, Suarez-Nieto MV, Sarno J, Nyame K, Xiong J, Pimentel Vera LN, Arozqueta Basurto J, Corbo M, Limaye A, Davis KL, Abu-Remaileh M, Gomez-Ospina N. CNS-wide repopulation by hematopoietic-derived microglia-like cells corrects progranulin deficiency in mice. Nat Commun 2024; 15:5654. [PMID: 38969669 PMCID: PMC11226701 DOI: 10.1038/s41467-024-49908-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Accepted: 06/17/2024] [Indexed: 07/07/2024] Open
Abstract
Hematopoietic stem cell transplantation can deliver therapeutic proteins to the central nervous system (CNS) through transplant-derived microglia-like cells. However, current conditioning approaches result in low and slow engraftment of transplanted cells in the CNS. Here we optimized a brain conditioning regimen that leads to rapid, robust, and persistent microglia replacement without adverse effects on neurobehavior or hematopoiesis. This regimen combines busulfan myeloablation and six days of Colony-stimulating factor 1 receptor inhibitor PLX3397. Single-cell analyses revealed unappreciated heterogeneity of microglia-like cells with most cells expressing genes characteristic of homeostatic microglia, brain-border-associated macrophages, and unique markers. Cytokine analysis in the CNS showed transient inductions of myeloproliferative and chemoattractant cytokines that help repopulate the microglia niche. Bone marrow transplant of progranulin-deficient mice conditioned with busulfan and PLX3397 restored progranulin in the brain and eyes and normalized brain lipofuscin storage, proteostasis, and lipid metabolism. This study advances our understanding of CNS repopulation by hematopoietic-derived cells and demonstrates its therapeutic potential for treating progranulin-dependent neurodegeneration.
Collapse
Affiliation(s)
- Pasqualina Colella
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| | - Ruhi Sayana
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
| | | | - Jolanda Sarno
- Hematology, Oncology, Stem Cell Transplant, and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
- Tettamanti Center, Fondazione IRCCS San Gerardo dei Tintori, 20900, Monza, Italy
| | - Kwamina Nyame
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA, 94305, USA
| | - Jian Xiong
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA, 94305, USA
| | | | | | - Marco Corbo
- MedGenome, Inc, 348 Hatch Dr, Foster City, CA, 94404, USA
| | - Anay Limaye
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA
- MedGenome, Inc, 348 Hatch Dr, Foster City, CA, 94404, USA
| | - Kara L Davis
- Hematology, Oncology, Stem Cell Transplant, and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, CA, 94305, USA
| | - Monther Abu-Remaileh
- Department of Chemical Engineering, Stanford University, Stanford, CA, 94305, USA
- Department of Genetics, Stanford University, Stanford, CA, 94305, USA
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, CA, 94305, USA
| | - Natalia Gomez-Ospina
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305, USA.
| |
Collapse
|
3
|
Reich M, Simon MJ, Polke B, Paris I, Werner G, Schrader C, Spieth L, Davis SS, Robinson S, de Melo GL, Schlaphoff L, Buschmann K, Berghoff S, Logan T, Nuscher B, de Weerd L, Edbauer D, Simons M, Suh JH, Sandmann T, Kariolis MS, DeVos SL, Lewcock JW, Paquet D, Capell A, Di Paolo G, Haass C. Peripheral expression of brain-penetrant progranulin rescues pathologies in mouse models of frontotemporal lobar degeneration. Sci Transl Med 2024; 16:eadj7308. [PMID: 38838131 DOI: 10.1126/scitranslmed.adj7308] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2023] [Accepted: 05/10/2024] [Indexed: 06/07/2024]
Abstract
Progranulin (PGRN) haploinsufficiency is a major risk factor for frontotemporal lobar degeneration with TAR DNA-binding protein 43 (TDP-43) pathology (FTLD-GRN). Multiple therapeutic strategies are in clinical development to restore PGRN in the CNS, including gene therapy. However, a limitation of current gene therapy approaches aimed to alleviate FTLD-associated pathologies may be their inefficient brain exposure and biodistribution. We therefore developed an adeno-associated virus (AAV) targeting the liver (L) to achieve sustained peripheral expression of a transferrin receptor (TfR) binding, brain-penetrant (b) PGRN variant [AAV(L):bPGRN] in two mouse models of FTLD-GRN, namely, Grn knockout and GrnxTmem106b double knockout mice. This therapeutic strategy avoids potential safety and biodistribution issues of CNS-administered AAVs and maintains sustained concentrations of PGRN in the brain after a single dose. AAV(L):bPGRN treatment reduced several FTLD-GRN-associated pathologies including severe motor function deficits, aberrant TDP-43 phosphorylation, dysfunctional protein degradation, lipid metabolism, gliosis, and neurodegeneration in the brain. The potential translatability of our findings was tested in an in vitro model using cocultured human induced pluripotent stem cell (hiPSC)-derived microglia lacking PGRN and TMEM106B and wild-type hiPSC-derived neurons. As in mice, aberrant TDP-43, lysosomal dysfunction, and neuronal loss were ameliorated after treatment with exogenous TfR-binding protein transport vehicle fused to PGRN (PTV:PGRN). Together, our studies suggest that peripherally administered brain-penetrant PGRN replacement strategies ameliorate FTLD-GRN relevant phenotypes including TDP-43 pathology, neurodegeneration, and behavioral deficits. Our data provide preclinical proof of concept for the use of this AAV platform for treatment of FTLD-GRN and potentially other CNS disorders.
Collapse
Affiliation(s)
- Marvin Reich
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), LMU Munich, 82152 Planegg, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
| | - Matthew J Simon
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Beate Polke
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Iñaki Paris
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Georg Werner
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Christian Schrader
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Lena Spieth
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| | - Sonnet S Davis
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Sophie Robinson
- Graduate School of Systemic Neurosciences (GSN), LMU Munich, 82152 Planegg, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | | | - Lennart Schlaphoff
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Graduate School of Systemic Neurosciences (GSN), LMU Munich, 82152 Planegg, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany
| | - Katrin Buschmann
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Stefan Berghoff
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany
- Max Planck Institute for Multidisciplinary Sciences, 37077 Göttingen, Germany
| | - Todd Logan
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Brigitte Nuscher
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | - Lis de Weerd
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
| | - Dieter Edbauer
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Munich Cluster for Systems Neurology (Synergy), 81377 Munich, Germany
| | - Mikael Simons
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
- Institute of Neuronal Cell Biology, Technical University Munich, 80802 Munich, Germany
- Munich Cluster for Systems Neurology (Synergy), 81377 Munich, Germany
| | - Jung H Suh
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | - Thomas Sandmann
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | | | - Sarah L DeVos
- Denali Therapeutics Inc., South San Francisco, CA 94080, USA
| | | | - Dominik Paquet
- Institute for Stroke and Dementia Research (ISD), University Hospital, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (Synergy), 81377 Munich, Germany
| | - Anja Capell
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
| | | | - Christian Haass
- German Center for Neurodegenerative Diseases (DZNE), 81377 Munich, Germany
- Metabolic Biochemistry, Biomedical Centre (BMC), Faculty of Medicine, LMU Munich, 81377 Munich, Germany
- Munich Cluster for Systems Neurology (Synergy), 81377 Munich, Germany
| |
Collapse
|
4
|
Cooper O, Hallett P, Isacson O. Upstream lipid and metabolic systems are potential causes of Alzheimer's disease, Parkinson's disease and dementias. FEBS J 2024; 291:632-645. [PMID: 36165619 PMCID: PMC10040476 DOI: 10.1111/febs.16638] [Citation(s) in RCA: 7] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2022] [Revised: 09/02/2022] [Accepted: 09/26/2022] [Indexed: 11/28/2022]
Abstract
Brain health requires circuits, cells and molecular pathways to adapt when challenged and to promptly reset once the challenge has resolved. Neurodegeneration occurs when adaptability becomes confined, causing challenges to overwhelm neural circuitry. Studies of rare and common neurodegenerative diseases suggest that the accumulation of lipids can compromise circuit adaptability. Using microglia as an example, we review data that suggest increased lipid concentrations cause dysfunctional inflammatory responses to immune challenges, leading to Alzheimer's disease, Parkinson's disease and dementia. We highlight current approaches to treat lipid metabolic and clearance pathways and identify knowledge gaps towards restoring adaptive homeostasis in individuals who are at-risk of losing cognition.
Collapse
Affiliation(s)
- Oliver Cooper
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478
| | - Penny Hallett
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478
| | - Ole Isacson
- Neuroregeneration Research Institute, McLean Hospital/Harvard Medical School, 115 Mill Street, Belmont, MA 02478
| |
Collapse
|
5
|
Ducharme S, Pijnenburg Y, Rohrer JD, Huey E, Finger E, Tatton N. Identifying and Diagnosing TDP-43 Neurodegenerative Diseases in Psychiatry. Am J Geriatr Psychiatry 2024; 32:98-113. [PMID: 37741764 PMCID: PMC11270911 DOI: 10.1016/j.jagp.2023.08.017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/19/2023] [Revised: 08/02/2023] [Accepted: 08/24/2023] [Indexed: 09/25/2023]
Abstract
Neuropsychiatric symptoms (NPS) are common manifestations of neurodegenerative disorders and are often early signs of those diseases. Among those neurodegenerative diseases, TDP-43 proteinopathies are an increasingly recognized cause of early neuropsychiatric manifestations. TDP-43-related diseases include frontotemporal dementia (FTD), amyotrophic lateral sclerosis (ALS), and Limbic-Predominant Age-Related TDP-43 Encephalopathy (LATE). The majority of TDP-43-related diseases are sporadic, but a significant proportion is hereditary, with progranulin (GRN) mutations and C9orf72 repeat expansions as the most common genetic etiologies. Studies reveal that NPS can be the initial manifestation of those diseases or can complicate disease course, but there is a lack of awareness among clinicians about TDP-43-related diseases, which leads to common diagnostic mistakes or delays. There is also emerging evidence that TDP-43 accumulations could play a role in late-onset primary psychiatric disorders. In the absence of robust biomarkers for TDP-43, the diagnosis remains primarily based on clinical assessment and neuroimaging. Given the association with psychiatric symptoms, clinical psychiatrists have a key role in the early identification of patients with TDP-43-related diseases. This narrative review provides a comprehensive overview of the pathobiology of TDP-43, resulting clinical presentations, and associated neuropsychiatric manifestations to help guide clinical practice.
Collapse
Affiliation(s)
- Simon Ducharme
- Department of Psychiatry (SD), Douglas Mental Health University Institute, McGill University, Montreal, Canada; McConnell Brain Imaging Centre, Montreal Neurological Institute, McGill University, Montreal, Canada.
| | - Yolande Pijnenburg
- Alzheimer Center Amsterdam, Department of Neurology, Amsterdam Neuroscience (YP), Vrije Universiteit Amsterdam, Amsterdam UMC, Amsterdam, The Netherlands
| | - Jonathan D Rohrer
- Dementia Research Centre, Department of Neurodegenerative Disease (JDR), UCL Queen Square Institute of Neurology, London, UK
| | - Edward Huey
- Taub Institute for Research on Alzheimer's Disease and the Aging Brain, Department of Psychiatry (EH), Columbia University, New York, NY
| | - Elizabeth Finger
- London Health Sciences Centre Parkwood Institute (EF), London, ON, Canada
| | | |
Collapse
|
6
|
McElhanon KE, Huff TC, Hirenallur-Shanthappa D, Miller RA, Christoforou N. Increased circulating progranulin is not sufficient to induce cardiac dysfunction or supraventricular arrhythmia. Sci Rep 2023; 13:21541. [PMID: 38057339 PMCID: PMC10700350 DOI: 10.1038/s41598-023-47311-5] [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: 04/14/2023] [Accepted: 11/12/2023] [Indexed: 12/08/2023] Open
Abstract
Atrial fibrillation (AF) is the most prevalent cardiac arrhythmia, and the incidence of new-onset AF has been increasing over the past two decades. Several factors contribute to the risk of developing AF including age, preexisting cardiovascular disease, chronic kidney disease, and obesity. Concurrent with the rise in AF, obesity has followed the same two-decade trend. The contribution of circulating proteins to obesity-related AF is of particular interest in the field. In this study, we investigated the effects of increased circulating levels of the glycoprotein progranulin on the development of supraventricular arrhythmias and changes to cardiac function. AAV8-mediated overexpression of full-length mouse progranulin was used to increase plasma protein levels and determine susceptibility to supraventricular arrhythmias and changes in cardiac structure and function. C57Bl/6N mice were subjected to increased circulating levels of progranulin for 20 weeks. Cardiac conduction was evaluated by surface ECG with and without isoproterenol challenge, and cardiac structure and function were measured by echocardiography after 20 weeks of circulating progranulin overexpression. Increased circulating levels of progranulin were maintained throughout the 20-week study. The cardiac structure and function remained unchanged in mice with increased circulating progranulin. ECG indices (P wave duration, P amplitude, QRS interval) were unaffected by increased progranulin levels and no arrhythmogenic events were observed following the isoproterenol challenge. In our model, increased levels of circulating progranulin were not sufficient to induce changes in cardiac structure and function or elicit ECG abnormalities suggestive of susceptibility to supraventricular arrhythmias.
Collapse
Affiliation(s)
- Kevin E McElhanon
- Rare Disease Research Unit, Worldwide Research, Development, and Medical, Pfizer, Inc., Cambridge, MA, USA
| | - Tyler C Huff
- Rare Disease Research Unit, Worldwide Research, Development, and Medical, Pfizer, Inc., Cambridge, MA, USA
| | | | - Russell A Miller
- Rare Disease Research Unit, Worldwide Research, Development, and Medical, Pfizer, Inc., Cambridge, MA, USA
| | - Nicolas Christoforou
- Rare Disease Research Unit, Worldwide Research, Development, and Medical, Pfizer, Inc., Cambridge, MA, USA.
| |
Collapse
|
7
|
Colella P, Sayana R, Suarez-Nieto MV, Sarno J, Nyame K, Xiong J, Vera LNP, Basurto JA, Corbo M, Limaye A, Davis KL, Abu-Remaileh M, Gomez-Ospina N. CNS Repopulation by Hematopoietic-Derived Microglia-Like Cells Corrects Progranulin deficiency. RESEARCH SQUARE 2023:rs.3.rs-3263412. [PMID: 37790525 PMCID: PMC10543302 DOI: 10.21203/rs.3.rs-3263412/v1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/05/2023]
Abstract
Hematopoietic stem cell transplantation can deliver therapeutic proteins to the CNS through donor-derived hematopoietic cells that become microglia-like cells. However, using standard conditioning approaches, hematopoietic stem cell transplantation is currently limited by low and slow engraftment of microglia-like cells. We report an efficient conditioning regimen based on Busulfan and a six-day course of microglia depletion using the colony-stimulating factor receptor 1 inhibitor PLX3397. Combining Busulfan-myeloablation and transient microglia depletion results in robust, rapid, and persistent microglia replacement by bone marrow-derived microglia-like cells throughout the CNS. Adding PLX3397 does not affect neurobehavior or has adverse effects on hematopoietic reconstitution. Through single-cell RNA sequencing and high-dimensional CyTOF mass cytometry, we show that microglia-like cells are a heterogeneous population and describe six distinct subpopulations. Though most bone-marrow-derived microglia-like cells can be classified as homeostatic microglia, their gene signature is a hybrid of homeostatic/embryonic microglia and border associated-macrophages. Busulfan-myeloablation and transient microglia depletion induce specific cytokines in the brain, ultimately combining myeloid proliferative and chemo-attractive signals that act locally to repopulate microglia from outside the niche. Importantly, this conditioning approach demonstrates therapeutic efficacy in a mouse model of GRN deficiency. Transplanting wild-type bone marrow into Grn-/- mice conditioned with Busulfan plus PLX3397 results in high engraftment of microglia-like cells in the brain and retina, restoring GRN levels and normalizing lipid metabolism.
Collapse
Affiliation(s)
- Pasqualina Colella
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305
| | - Ruhi Sayana
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305
| | | | - Jolanda Sarno
- Hematology, Oncology, Stem Cell Transplant, and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, CA, 94305
| | - Kwamina Nyame
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305
- Department of Genetics, Stanford University, Stanford, CA 94305
| | - Jian Xiong
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305
- Department of Genetics, Stanford University, Stanford, CA 94305
| | | | | | - Marco Corbo
- MedGenome, Inc, 348 Hatch Dr, Foster City, CA 94404
| | - Anay Limaye
- MedGenome, Inc, 348 Hatch Dr, Foster City, CA 94404
| | - Kara Lynn Davis
- Hematology, Oncology, Stem Cell Transplant, and Regenerative Medicine, Department of Pediatrics, Stanford University, Stanford, CA, 94305
| | - Monther Abu-Remaileh
- Department of Chemical Engineering, Stanford University, Stanford, CA 94305
- Department of Genetics, Stanford University, Stanford, CA 94305
- The Institute for Chemistry, Engineering and Medicine for Human Health (Sarafan ChEM-H), Stanford University, Stanford, USA
| | - Natalia Gomez-Ospina
- Department of Pediatrics, Stanford University School of Medicine, Stanford, CA, 94305
| |
Collapse
|
8
|
Feng T, Minevich G, Liu P, Qin HX, Wozniak G, Pham J, Pham K, Korgaonkar A, Kurnellas M, Defranoux NA, Long H, Mitra A, Hu F. AAV- GRN partially corrects motor deficits and ALS/FTLD-related pathology in Tmem106b-/-Grn-/- mice. iScience 2023; 26:107247. [PMID: 37519899 PMCID: PMC10371829 DOI: 10.1016/j.isci.2023.107247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2023] [Revised: 05/18/2023] [Accepted: 06/26/2023] [Indexed: 08/01/2023] Open
Abstract
Loss of function of progranulin (PGRN), encoded by the granulin (GRN) gene, is implicated in several neurodegenerative diseases. Several therapeutics to boost PGRN levels are currently in clinical trials. However, it is difficult to test the efficacy of PGRN-enhancing drugs in mouse models due to the mild phenotypes of Grn-/- mice. Recently, mice deficient in both PGRN and TMEM106B were shown to develop severe motor deficits and pathology. Here, we show that intracerebral ventricle injection of PGRN-expressing AAV1/9 viruses partially rescues motor deficits, neuronal loss, glial activation, and lysosomal abnormalities in Tmem106b-/-Grn-/- mice. Widespread expression of PGRN is detected in both the brain and spinal cord for both AAV subtypes. However, AAV9 but not AAV1-mediated expression of PGRN results in high levels of PGRN in the serum. Together, these data support using the Tmem106b-/-Grn-/- mouse strain as a robust mouse model to determine the efficacy of PGRN-elevating therapeutics.
Collapse
Affiliation(s)
- Tuancheng Feng
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | | | - Pengan Liu
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | - Henry Xin Qin
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| | | | - Jenny Pham
- Alector Inc, South San Francisco, CA 94080, USA
| | - Khanh Pham
- Alector Inc, South San Francisco, CA 94080, USA
| | | | | | | | - Hua Long
- Alector Inc, South San Francisco, CA 94080, USA
| | | | - Fenghua Hu
- Department of Molecular Biology and Genetics, Weill Institute for Cell and Molecular Biology, Cornell University, Ithaca, NY 14853, USA
| |
Collapse
|
9
|
Simon MJ, Logan T, DeVos SL, Di Paolo G. Lysosomal functions of progranulin and implications for treatment of frontotemporal dementia. Trends Cell Biol 2023; 33:324-339. [PMID: 36244875 DOI: 10.1016/j.tcb.2022.09.006] [Citation(s) in RCA: 35] [Impact Index Per Article: 35.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/25/2022] [Revised: 09/15/2022] [Accepted: 09/15/2022] [Indexed: 12/12/2022]
Abstract
Loss-of-function heterozygous mutations in GRN, the gene encoding progranulin (PGRN), were identified in patients with frontotemporal lobar degeneration (FTLD) almost two decades ago and are generally linked to reduced PGRN protein expression levels. Although initial characterization of PGRN function primarily focused on its role in extracellular signaling as a secreted protein, more recent studies revealed critical roles of PGRN in regulating lysosome function, including proteolysis and lipid degradation, consistent with its lysosomal localization. Emerging from these studies is the notion that PGRN regulates glucocerebrosidase activity via direct chaperone activities and via interaction with prosaposin (i.e., a key regulator of lysosomal sphingolipid-metabolizing enzymes), as well as with the anionic phospholipid bis(monoacylglycero)phosphate. This emerging lysosomal biology of PGRN identified novel and promising opportunities in therapeutic discovery as well as biomarker development.
Collapse
Affiliation(s)
| | - Todd Logan
- Denali Therapeutics, South San Francisco, CA, USA
| | | | | |
Collapse
|
10
|
Kashyap SN, Boyle NR, Roberson ED. Preclinical Interventions in Mouse Models of Frontotemporal Dementia Due to Progranulin Mutations. Neurotherapeutics 2023; 20:140-153. [PMID: 36781744 PMCID: PMC10119358 DOI: 10.1007/s13311-023-01348-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 01/20/2023] [Indexed: 02/15/2023] Open
Abstract
Heterozygous loss-of-function mutations in progranulin (GRN) cause frontotemporal dementia (FTD), a leading cause of early-onset dementia characterized clinically by behavioral, social, and language deficits. There are currently no FDA-approved therapeutics for FTD-GRN, but this has been an active area of investigation, and several approaches are now in clinical trials. Here, we review preclinical development of therapies for FTD-GRN with a focus on testing in mouse models. Since most FTD-GRN-associated mutations cause progranulin haploinsufficiency, these approaches focus on raising progranulin levels. We begin by considering the disorders associated with altered progranulin levels, and then review the basics of progranulin biology including its lysosomal, neurotrophic, and immunomodulatory functions. We discuss mouse models of progranulin insufficiency and how they have been used in preclinical studies on a variety of therapeutic approaches. These include approaches to raise progranulin expression from the normal allele or facilitate progranulin production by the mutant allele, as well as approaches to directly increase progranulin levels by delivery across the blood-brain barrier or by gene therapy. Several of these approaches have entered clinical trials, providing hope that new therapies for FTD-GRN may be the next frontier in the treatment of neurodegenerative disease.
Collapse
Affiliation(s)
- Shreya N Kashyap
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Medical Scientist Training Program, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Nicholas R Boyle
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Medical Scientist Training Program, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA
| | - Erik D Roberson
- Center for Neurodegeneration and Experimental Therapeutics, Alzheimer's Disease Center, Medical Scientist Training Program, Department of Neurology, University of Alabama at Birmingham, Birmingham, AL, 35294, USA.
| |
Collapse
|
11
|
Colon-Moran W, Baer A, Lamture G, Stapleton JT, Fischer JW, Bhattarai N. A short hepatitis C virus NS5A peptide expression by AAV vector modulates human T cell activation and reduces vector immunogenicity. Gene Ther 2022; 29:616-623. [PMID: 34759330 PMCID: PMC9091046 DOI: 10.1038/s41434-021-00302-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2021] [Revised: 10/22/2021] [Accepted: 10/26/2021] [Indexed: 01/09/2023]
Abstract
Viral vector-mediated gene therapies have the potential to treat many human diseases; however, host immune responses against the vector and/or the transgene pose a safety risk to the patients and can negatively impact product efficacy. Thus, novel strategies to reduce vector immunogenicity are critical for the advancement of these therapies. T cell activation (TCA) is required for the development of immune responses during gene therapy. We hypothesized that modulation of TCA by incorporating a novel viral immunomodulatory factor into a viral vector may reduce unwanted TCA and immune responses during gene therapy. To test this hypothesis, we identified an immunomodulatory domain of the hepatitis C virus (HCV) NS protein 5A (NS5A) protein and studied the effect of viral vectors expressing NS5A peptide on TCA. Lentiviral vector-mediated expression of a short 20-mer peptide derived from the NS5A protein in human T cells was sufficient to inhibit TCA. Synthetic 20-mer NS5A peptide also inhibited TCA in primary human T cells. Mechanistically, the NS5A protein interacted with Lck and inhibited proximal TCR signaling. Importantly, NS5A peptide expression did not cause global T cell signaling dysfunction as distal T cell signaling was not inhibited. Finally, recombinant adeno-associated virus (AAV) vector expressing the 20-mer NS5A peptide reduced both the recall antigen and the TCR-mediated activation of human T cells and did not cause global T cell signaling dysfunction. Together, these data suggest that expression of a 20-mer NS5A peptide by an AAV vector may reduce unwanted TCA and may contribute to lower vector immunogenicity during gene therapy.
Collapse
Affiliation(s)
- Winston Colon-Moran
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Alan Baer
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
| | - Gauri Lamture
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
- Adicet Bio, Inc., Menlo Park, CA, USA
| | - Jack T Stapleton
- Research Service, Iowa City Veterans Affairs Medical Center, Iowa City, IA, USA
- Departments of Internal Medicine and Microbiology, University of Iowa, Iowa City, IA, USA
| | - Joseph W Fischer
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA
- AstraZeneca, Gaithersburg, MD, USA
| | - Nirjal Bhattarai
- Division of Cellular and Gene Therapies, Office of Tissues and Advanced Therapies, Center for Biologics Evaluation and Research, Food and Drug Administration, Silver Spring, MD, 20993, USA.
| |
Collapse
|
12
|
Kofoed RH, Dibia CL, Noseworthy K, Xhima K, Vacaresse N, Hynynen K, Aubert I. Efficacy of gene delivery to the brain using AAV and ultrasound depends on serotypes and brain areas. J Control Release 2022; 351:667-680. [DOI: 10.1016/j.jconrel.2022.09.048] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 09/18/2022] [Accepted: 09/23/2022] [Indexed: 02/01/2023]
|
13
|
Neuroimmune dysfunction in frontotemporal dementia: Insights from progranulin and C9orf72 deficiency. Curr Opin Neurobiol 2022; 76:102599. [PMID: 35792478 PMCID: PMC9798541 DOI: 10.1016/j.conb.2022.102599] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2022] [Revised: 05/25/2022] [Accepted: 05/31/2022] [Indexed: 12/31/2022]
Abstract
Neuroimmune dysfunction is a cardinal feature of neurodegenerative diseases. But how immune dysregulation in the brain and peripheral organs contribute to neurodegeneration remains unclear. Here, we discuss the recent advances highlighting neuroimmune dysfunction as a key disease-driving factor in frontotemporal dementia (FTD). We provide an overview of the clinical observations supporting a high prevalence of autoimmune diseases in FTD patients with mutations in GRN or C9orf72. We then focus on a myriad of evidence from human genetic studies, mouse models, in vitro assays, and multi-omics platform, which indicate that haploinsufficiency in GRN and C9orf72 promotes neuroimmune dysfunction and contributes to neurodegeneration and premature death. These compelling data provide key insights to disease mechanisms, biomarker discovery, and therapeutic interventions for FTD (120 words).
Collapse
|
14
|
Zhang Y, Xing CJ, Liu X, Li YH, Jia J, Feng JG, Yang CJ, Chen Y, Zhou J. Thioredoxin-Interacting Protein (TXNIP) Knockdown Protects against Sepsis-Induced Brain Injury and Cognitive Decline in Mice by Suppressing Oxidative Stress and Neuroinflammation. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8645714. [PMID: 35571246 PMCID: PMC9098358 DOI: 10.1155/2022/8645714] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/19/2021] [Revised: 04/17/2022] [Accepted: 04/23/2022] [Indexed: 11/19/2022]
Abstract
Sepsis-associated encephalopathy (SAE) is linked to increased morbidity and mortality rates in patients with sepsis. Increased cytokine production and neuronal apoptosis are implicated in the pathogenesis of the SAE. Neuroinflammation plays a major role in sepsis-induced brain injury. Thioredoxin-interacting protein (TXNIP), an inhibitor of thioredoxin, is associated with oxidative stress and inflammation. However, whether the TXNIP is involved in the sepsis-induced brain injury and the underlying mechanism is yet to be elucidated. Therefore, the present study was aimed at elucidating the effects of TXNIP knockdown on sepsis-induced brain injury and cognitive decline in mice. Lipopolysaccharide (LPS) was injected intraperitoneally to induce sepsis brain injury in mice. The virus-carrying control or TXNIP shRNA was injected into the lateral ventricle of the brain 4 weeks before the LPS treatment. The histological changes in the hippocampal tissues, encephaledema, and cognitive function were detected, respectively. Also, the 7-day survival rate was recorded. Furthermore, the alterations in microglial activity, oxidative response, proinflammatory factors, apoptosis, protein levels (TXNIP and NLRP3 inflammasome), and apoptosis were examined in the hippocampal tissues. The results demonstrated that the TXNIP and NLRP3 inflammasome expression levels were increased at 6, 12, and 24 h post-LPS injection. TXNIP knockdown dramatically ameliorated the 7-day survival rate, cognitive decline, brain damage, neuronal apoptosis, and the brain water content, inhibited the activation of microglia, downregulated the NLRP3/caspase-1 signaling pathway, and reduced the oxidative stress and the neuroinflammatory cytokine levels at 24 h post-LPS injection. These results suggested a crucial effect of TXNIP knockdown on the mechanism of brain injury and cognitive decline in sepsis mice via suppressing oxidative stress and neuroinflammation. Thus, TXNIP might be a potential therapeutic target for SAE patients.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Cheng-Jun Xing
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Xiao Liu
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Ya-Hong Li
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jing Jia
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Anesthesiology, Southwest Medical University, Luzhou, China
| | - Jian-Guo Feng
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Anesthesiology, Southwest Medical University, Luzhou, China
| | - Cheng-Jie Yang
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Anesthesiology, Southwest Medical University, Luzhou, China
| | - Ye Chen
- Department of Traditional Chinese Medicine, The Affiliated Hospital of Southwest Medical University, Luzhou, China
| | - Jun Zhou
- Department of Anesthesiology, The Affiliated Hospital of Southwest Medical University, Luzhou, China
- Laboratory of Anesthesiology, Southwest Medical University, Luzhou, China
| |
Collapse
|
15
|
Tanaka Y, Kusumoto SY, Honma Y, Takeya K, Eto M. Overexpression of progranulin increases pathological protein accumulation by suppressing autophagic flux. Biochem Biophys Res Commun 2022; 611:78-84. [PMID: 35483222 DOI: 10.1016/j.bbrc.2022.04.064] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Accepted: 04/13/2022] [Indexed: 01/01/2023]
Abstract
Progranulin (PGRN) haploinsufficiency from autosomal dominant mutations in the PGRN gene causes frontotemporal lobar degeneration, which is characterized by cytoplasmic inclusions predominantly containing TDP-43 (FTLD-TDP). PGRN supplementation for patients with a PGRN gene mutation has recently been proposed as a therapeutic strategy to suppress FTLD-TDP. However, it currently remains unclear whether excessive amounts of PGRN are beneficial or harmful. We herein report the effects of PGRN overexpression on autophagic flux in a cultured cell model. PGRN overexpression increased the level of an autophagosome marker without promoting autophagosome formation and decreased the signal intensity of an autolysosome marker, indicating the suppression of autophagic flux due to reductions in the formation of autolysosomes. Assessments of lysosome numbers and biogenesis using LysoTracker and cells stably expressing TFEB-GFP, respectively, indicated that PGRN overexpression increased the lysosome numbers without lysosomal biogenesis. These results suggest that PGRN overexpression suppressed autophagic flux by inhibiting autophagosome-lysosome fusion. Moreover, PGRN overexpression enhanced polyglutamine aggregation and aggregate-prone TDP-43 accumulation, indicating that the suppression of autophagic flux by excessive amounts of PGRN worsens the pathology of neurodegenerative diseases.
Collapse
Affiliation(s)
- Yoshinori Tanaka
- Biochemistry Unit, Faculty of Veterinary Medicine, Okayama University of Science, Imabari-shi, Ehime, Japan.
| | - Shun-Ya Kusumoto
- Biochemistry Unit, Faculty of Veterinary Medicine, Okayama University of Science, Imabari-shi, Ehime, Japan
| | - Yuki Honma
- Biochemistry Unit, Faculty of Veterinary Medicine, Okayama University of Science, Imabari-shi, Ehime, Japan
| | - Kosuke Takeya
- Biochemistry Unit, Faculty of Veterinary Medicine, Okayama University of Science, Imabari-shi, Ehime, Japan
| | - Masumi Eto
- Biochemistry Unit, Faculty of Veterinary Medicine, Okayama University of Science, Imabari-shi, Ehime, Japan
| |
Collapse
|
16
|
Kofoed RH, Heinen S, Silburt J, Dubey S, Dibia CL, Maes M, Simpson EM, Hynynen K, Aubert I. Transgene distribution and immune response after ultrasound delivery of rAAV9 and PHP.B to the brain in a mouse model of amyloidosis. Mol Ther Methods Clin Dev 2021; 23:390-405. [PMID: 34761053 PMCID: PMC8560718 DOI: 10.1016/j.omtm.2021.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Revised: 08/12/2021] [Accepted: 10/05/2021] [Indexed: 01/01/2023]
Abstract
Efficient disease-modifying treatments for Alzheimer disease, the most common form of dementia, have yet to be established. Gene therapy has the potential to provide the long-term production of therapeutic in the brain following a single administration. However, the blood-brain barrier poses a challenge for gene delivery to the adult brain. We investigated the transduction efficiency and immunological response following non-invasive gene-delivery strategies to the brain of a mouse model of amyloidosis. Two emerging technologies enabling gene delivery across the blood-brain barrier were used to establish the minimal vector dosage required to reach the brain: (1) focused ultrasound combined with intravenous microbubbles, which increases the permeability of the blood-brain barrier at targeted sites and (2) the recombinant adeno-associated virus (rAAV)-based capsid named rAAV-PHP.B. We found that equal intravenous dosages of rAAV9 combined with focused ultrasound, or rAAV-PHP.B, were required for brain gene delivery. In contrast to rAAV9, focused ultrasound did not decrease the rAAV-PHP.B dosage required to transduce brain cells in a mouse model of amyloidosis. The non-invasive rAAV delivery to the brain using rAAV-PHP.B or rAAV9 with focused ultrasound triggered an immune reaction including major histocompatibility complex class II expression, complement system and microglial activation, and T cell infiltration.
Collapse
Affiliation(s)
- Rikke Hahn Kofoed
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Stefan Heinen
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Joseph Silburt
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Sonam Dubey
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Chinaza Lilian Dibia
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Miriam Maes
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| | - Elizabeth M. Simpson
- Centre for Molecular Medicine and Therapeutics at British Columbia Children’s Hospital, Department of Medical Genetics, The University of British Columbia, Vancouver, BC V5Z 4H4, Canada
| | - Kullervo Hynynen
- Physical Sciences, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Medical Biophysics, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5G 1L7, Canada
| | - Isabelle Aubert
- Biological Sciences, Hurvitz Brain Sciences Research Program, Sunnybrook Research Institute, Sunnybrook Health Sciences Centre, Toronto, ON M4N 3M5, Canada
- Department of Laboratory Medicine and Pathobiology, Temerty Faculty of Medicine, University of Toronto, Toronto, ON M5S 1A8, Canada
| |
Collapse
|
17
|
Morris JA, Boshoff CH, Schor NF, Wong LM, Gao G, Davidson BL. Next-generation strategies for gene-targeted therapies of central nervous system disorders: A workshop summary. Mol Ther 2021; 29:3332-3344. [PMID: 34547463 DOI: 10.1016/j.ymthe.2021.09.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2021] [Revised: 09/13/2021] [Accepted: 09/14/2021] [Indexed: 12/30/2022] Open
Abstract
The National Institute of Neurological Disorders and Stroke (NINDS) held a workshop titled "Next generation strategies for gene-targeted therapies of central nervous system (CNS) disorders" in September 2019 in Bethesda, MD, USA. The meeting brought together a multi-disciplinary group of experts in the field of CNS-directed gene-targeted therapy delivery from academia, industry, advocacy, and the government. The group was charged with identifying the key challenges and gaps in this evolving field, as well as suggesting potential solutions. The workshop was divided into four sessions: (1) control of level and location, (2) improving delivery and distribution, (3) enhancing models and manufacturing, and (4) impacting patients. Prior to the workshop, NINDS established working groups of key opinion leaders (KOLs) for each session. In pre-meeting teleconferences, KOLs were tasked with identifying the research gaps and key obstacles that delay and/or prevent gene-targeted therapies to move into the clinic. This approach allowed for the workshop to begin with problem-solving discussions and strategy development, as the key issues had been established. The overall purpose of the workshop was to consider knowledge gaps and potential strategies to inform the community around CNS gene-targeted therapies, including but not limited to researchers and funders.
Collapse
Affiliation(s)
- Jill A Morris
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA.
| | - Chris H Boshoff
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Nina F Schor
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Ling M Wong
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, MD 20892, USA
| | - Guangping Gao
- Horae Gene Therapy Center, UMass Medical School, Worcester, MA 01605, USA
| | - Beverly L Davidson
- Children's Hospital of Philadelphia and University of Pennsylvania, Philadelphia, PA 19104, USA.
| |
Collapse
|
18
|
Schumann L, Wilken-Schmitz A, Trautmann S, Vogel A, Schreiber Y, Hahnefeld L, Gurke R, Geisslinger G, Tegeder I. Increased Fat Taste Preference in Progranulin-Deficient Mice. Nutrients 2021; 13:4125. [PMID: 34836380 PMCID: PMC8623710 DOI: 10.3390/nu13114125] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2021] [Revised: 10/22/2021] [Accepted: 11/12/2021] [Indexed: 11/25/2022] Open
Abstract
Progranulin deficiency in mice is associated with deregulations of the scavenger receptor signaling of CD36/SCARB3 in immune disease models, and CD36 is a dominant receptor in taste bud cells in the tongue and contributes to the sensation of dietary fats. Progranulin-deficient mice (Grn-/-) are moderately overweight during middle age. We therefore asked if there was a connection between progranulin/CD36 in the tongue and fat taste preferences. By using unbiased behavioral analyses in IntelliCages and Phenomaster cages we showed that progranulin-deficient mice (Grn-/-) developed a strong preference of fat taste in the form of 2% milk over 0.3% milk, and for diluted MCTs versus tap water. The fat preference in the 7d-IntelliCage observation period caused an increase of 10% in the body weight of Grn-/- mice, which did not occur in the wildtype controls. CD36 expression in taste buds was reduced in Grn-/- mice at RNA and histology levels. There were no differences in the plasma or tongue lipids of various classes including sphingolipids, ceramides and endocannabinoids. The data suggest that progranulin deficiency leads to a lower expression of CD36 in the tongue resulting in a stronger urge for fatty taste and fatty nutrition.
Collapse
Affiliation(s)
- Lana Schumann
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
| | - Annett Wilken-Schmitz
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
| | - Sandra Trautmann
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
| | - Alexandra Vogel
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
| | - Yannick Schreiber
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
| | - Lisa Hahnefeld
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
| | - Robert Gurke
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
| | - Gerd Geisslinger
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
- Fraunhofer Institute for Translational Medicine and Pharmacology ITMP, 60596 Frankfurt, Germany
- Fraunhofer Cluster of Excellence for Immune Mediated Diseases (CIMD), 60596 Frankfurt, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Faculty of Medicine, Goethe-University Frankfurt, 60590 Frankfurt, Germany; (L.S.); (A.W.-S.); (S.T.); (A.V.); (Y.S.); (L.H.); (R.G.); (G.G.)
| |
Collapse
|
19
|
Level of hM4D(Gi) DREADD Expression Determines Inhibitory and Neurotoxic Effects in the Hippocampus. eNeuro 2021; 8:ENEURO.0105-21.2021. [PMID: 34620623 PMCID: PMC8570686 DOI: 10.1523/eneuro.0105-21.2021] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2021] [Revised: 08/16/2021] [Accepted: 08/20/2021] [Indexed: 12/12/2022] Open
Abstract
Selective neuromodulation using designer receptors exclusively activated by designer drugs (DREADDs) has become an increasingly important research tool, as well as an emerging therapeutic approach. However, the safety profile of DREADD expression is unknown. Here, different titers of adeno-associated viral (AAV) vector were administered in an attempt to vary total expression levels of the inhibitory DREADD hM4D(Gi) in excitatory hippocampal neurons. Male Sprague Dawley rats were injected with AAV2/7 encoding DREADD-mCherry, DREADD, or mCherry. Pronounced neuronal loss and neuroinflammatory reactions were observed after transduction with the high titer DREADD AAV, which also resulted in the highest DREADD expression levels. No such effects were observed in the mCherry control group, despite an equally high titer, nor in conditions where lower viral vector titers were injected. In the high titer DREADD conditions, dentate gyrus (DG) evoked potentials were inhibited on clozapine-induced activation of hM4D(Gi), while in low titer conditions DG evoked potentials were enhanced. Recordings of single neuronal activity nevertheless indicated a reduction in spontaneous firing of granule cell layer neurons. Our results indicate that prolonged, high levels of DREADD expression can have neurotoxic effects and that chemogenetic suppression of excitatory hippocampal neurons can paradoxically enhance DG evoked potentials.
Collapse
|
20
|
Jensen TL, Gøtzsche CR, Woldbye DPD. Current and Future Prospects for Gene Therapy for Rare Genetic Diseases Affecting the Brain and Spinal Cord. Front Mol Neurosci 2021; 14:695937. [PMID: 34690692 PMCID: PMC8527017 DOI: 10.3389/fnmol.2021.695937] [Citation(s) in RCA: 37] [Impact Index Per Article: 12.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2021] [Accepted: 09/02/2021] [Indexed: 12/12/2022] Open
Abstract
In recent years, gene therapy has been raising hopes toward viable treatment strategies for rare genetic diseases for which there has been almost exclusively supportive treatment. We here review this progress at the pre-clinical and clinical trial levels as well as market approvals within diseases that specifically affect the brain and spinal cord, including degenerative, developmental, lysosomal storage, and metabolic disorders. The field reached an unprecedented milestone when Zolgensma® (onasemnogene abeparvovec) was approved by the FDA and EMA for in vivo adeno-associated virus-mediated gene replacement therapy for spinal muscular atrophy. Shortly after EMA approved Libmeldy®, an ex vivo gene therapy with lentivirus vector-transduced autologous CD34-positive stem cells, for treatment of metachromatic leukodystrophy. These successes could be the first of many more new gene therapies in development that mostly target loss-of-function mutation diseases with gene replacement (e.g., Batten disease, mucopolysaccharidoses, gangliosidoses) or, less frequently, gain-of-toxic-function mutation diseases by gene therapeutic silencing of pathologic genes (e.g., amyotrophic lateral sclerosis, Huntington's disease). In addition, the use of genome editing as a gene therapy is being explored for some diseases, but this has so far only reached clinical testing in the treatment of mucopolysaccharidoses. Based on the large number of planned, ongoing, and completed clinical trials for rare genetic central nervous system diseases, it can be expected that several novel gene therapies will be approved and become available within the near future. Essential for this to happen is the in depth characterization of short- and long-term effects, safety aspects, and pharmacodynamics of the applied gene therapy platforms.
Collapse
Affiliation(s)
- Thomas Leth Jensen
- Department of Neurology, Rigshospitalet University Hospital, Copenhagen, Denmark
| | | | | |
Collapse
|
21
|
On the cusp of cures: Breakthroughs in Batten disease research. Curr Opin Neurobiol 2021; 72:48-54. [PMID: 34571324 DOI: 10.1016/j.conb.2021.08.003] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2021] [Revised: 08/12/2021] [Accepted: 08/17/2021] [Indexed: 12/27/2022]
Abstract
Batten disease is a family of rare, lysosomal disorders caused by mutations in one of at least 13 genes, which encode a diverse set of lysosomal and extralysosomal proteins. Despite decades of research, the development of effective therapies has remained intractable. But now, the field is experiencing rapid, unprecedented progress on multiple fronts. New tools are providing insights into previously unsolvable problems, with molecular functions now known for nine Batten disease proteins. Protein interactome data are uncovering potential functional overlap between several Batten disease proteins, providing long-sought links between seemingly disparate proteins. Understanding of cellular etiology is elucidating contributions from and interactions between various CNS cell types. Collectively, this explosion in insight is hastening an unparalleled period of therapeutic breakthroughs, with multiple therapies showing great promise in preclinical and clinical studies. The coming years will provide a continuation of this rapid progress, with the promise of effective treatments giving patients hope.
Collapse
|
22
|
Zin EA, Han D, Tran J, Morisson-Welch N, Visel M, Kuronen M, Flannery JG. Outcomes of progranulin gene therapy in the retina are dependent on time and route of delivery. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2021; 22:40-51. [PMID: 34485593 PMCID: PMC8390452 DOI: 10.1016/j.omtm.2021.05.009] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/05/2021] [Accepted: 05/19/2021] [Indexed: 11/25/2022]
Abstract
Neuronal ceroid lipofuscinosis (NCL) is a family of neurodegenerative diseases caused by mutations to genes related to lysosomal function. One variant, CNL11, is caused by mutations to the gene encoding the protein progranulin, which regulates neuronal lysosomal function. Absence of progranulin causes cerebellar atrophy, seizures, dementia, and vision loss. As progranulin gene therapies targeting the brain are developed, it is advantageous to focus on the retina, as its characteristics are beneficial for gene therapy development: the retina is easily visible through direct imaging, can be assessed through quantitative methods in vivo, and requires smaller amounts of adeno-associated virus (AAV). In this study we characterize the retinal degeneration in a progranulin knockout mouse model of CLN11 and study the effects of gene replacement at different time points. Mice heterologously expressing progranulin showed a reduction in lipofuscin deposits and microglia infiltration. While mice that receive systemic AAV92YF-scCAG-PGRN at post-natal day 3 or 4 show a reduction in retina thinning, mice injected intravitreally at months 1 and 6 with AAV2.7m8-scCAG-PGRN exhibit no improvement, and mice injected at 12 months of age have thinner retinas than do their controls. Thus, delivery of progranulin proves to be time sensitive and dependent on route of administration, requiring early delivery for optimal therapeutic benefit.
Collapse
Affiliation(s)
- Emilia A Zin
- Vision Science Group, School of Optometry, UC Berkeley, Berkeley, CA 94720, USA
| | - Daisy Han
- Department of Integrative Biology, UC Berkeley, Berkeley, CA 94720, USA
| | - Jennifer Tran
- School of Optometry, UC Berkeley, Berkeley, CA 94720, USA
| | | | - Meike Visel
- Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, CA 94720, USA
| | - Mervi Kuronen
- Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, CA 94720, USA
| | - John G Flannery
- Vision Science Group, School of Optometry, UC Berkeley, Berkeley, CA 94720, USA.,School of Optometry, UC Berkeley, Berkeley, CA 94720, USA.,Helen Wills Neuroscience Institute, UC Berkeley, Berkeley, CA 94720, USA
| |
Collapse
|
23
|
Regulated control of gene therapies by drug-induced splicing. Nature 2021; 596:291-295. [PMID: 34321659 PMCID: PMC8966400 DOI: 10.1038/s41586-021-03770-2] [Citation(s) in RCA: 65] [Impact Index Per Article: 21.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2020] [Accepted: 06/23/2021] [Indexed: 01/13/2023]
Abstract
So far, gene therapies have relied on complex constructs that cannot be finely controlled1,2. Here we report a universal switch element that enables precise control of gene replacement or gene editing after exposure to a small molecule. The small-molecule inducers are currently in human use, are orally bioavailable when given to animals or humans and can reach both peripheral tissues and the brain. Moreover, the switch system, which we denote Xon, does not require the co-expression of any regulatory proteins. Using Xon, the translation of the desired elements for controlled gene replacement or gene editing machinery occurs after a single oral dose of the inducer, and the robustness of expression can be controlled by the drug dose, protein stability and redosing. The ability of Xon to provide temporal control of protein expression can be adapted for cell-biology applications and animal studies. Additionally, owing to the oral bioavailability and safety of the drugs used, the Xon switch system provides an unprecedented opportunity to refine and tailor the application of gene therapies in humans.
Collapse
|
24
|
Terryn J, Verfaillie CM, Van Damme P. Tweaking Progranulin Expression: Therapeutic Avenues and Opportunities. Front Mol Neurosci 2021; 14:713031. [PMID: 34366786 PMCID: PMC8343103 DOI: 10.3389/fnmol.2021.713031] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2021] [Accepted: 07/07/2021] [Indexed: 01/01/2023] Open
Abstract
Frontotemporal dementia (FTD) is a neurodegenerative disease, leading to behavioral changes and language difficulties. Heterozygous loss-of-function mutations in progranulin (GRN) induce haploinsufficiency of the protein and are associated with up to one-third of all genetic FTD cases worldwide. While the loss of GRN is primarily associated with neurodegeneration, the biological functions of the secreted growth factor-like protein are more diverse, ranging from wound healing, inflammation, vasculogenesis, and metabolic regulation to tumor cell growth and metastasis. To date, no disease-modifying treatments exist for FTD, but different therapeutic approaches to boost GRN levels in the central nervous system are currently being developed (including AAV-mediated GRN gene delivery as well as anti-SORT1 antibody therapy). In this review, we provide an overview of the multifaceted regulation of GRN levels and the corresponding therapeutic avenues. We discuss the opportunities, advantages, and potential drawbacks of the diverse approaches. Additionally, we highlight the therapeutic potential of elevating GRN levels beyond patients with loss-of-function mutations in GRN.
Collapse
Affiliation(s)
- Joke Terryn
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium.,Laboratory of Neurobiology, Center for Brain and Disease Research, VIB, Leuven, Belgium
| | - Catherine M Verfaillie
- Department of Development and Regeneration, Interdepartmental Stem Cell Institute, KU Leuven-University of Leuven, Leuven, Belgium
| | - Philip Van Damme
- Department of Neurosciences, Experimental Neurology and Leuven Brain Institute (LBI), KU Leuven-University of Leuven, Leuven, Belgium.,Laboratory of Neurobiology, Center for Brain and Disease Research, VIB, Leuven, Belgium.,Department of Neurology, University Hospitals Leuven, Leuven, Belgium
| |
Collapse
|
25
|
Adeno-Associated Viral Vectors as Versatile Tools for Parkinson's Research, Both for Disease Modeling Purposes and for Therapeutic Uses. Int J Mol Sci 2021; 22:ijms22126389. [PMID: 34203739 PMCID: PMC8232322 DOI: 10.3390/ijms22126389] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 06/09/2021] [Accepted: 06/11/2021] [Indexed: 12/17/2022] Open
Abstract
It is without any doubt that precision medicine therapeutic strategies targeting neurodegenerative disorders are currently witnessing the spectacular rise of newly designed approaches based on the use of viral vectors as Trojan horses for the controlled release of a given genetic payload. Among the different types of viral vectors, adeno-associated viruses (AAVs) rank as the ones most commonly used for the purposes of either disease modeling or for therapeutic strategies. Here, we reviewed the current literature dealing with the use of AAVs within the field of Parkinson’s disease with the aim to provide neuroscientists with the advice and background required when facing a choice on which AAV might be best suited for addressing a given experimental challenge. Accordingly, here we will be summarizing some insights on different AAV serotypes, and which would be the most appropriate AAV delivery route. Next, the use of AAVs for modeling synucleinopathies is highlighted, providing potential readers with a landscape view of ongoing pre-clinical and clinical initiatives pushing forward AAV-based therapeutic approaches for Parkinson’s disease and related synucleinopathies.
Collapse
|
26
|
Vieira SRL, Morris HR. Neurodegenerative Disease Risk in Carriers of Autosomal Recessive Disease. Front Neurol 2021; 12:679927. [PMID: 34149605 PMCID: PMC8211888 DOI: 10.3389/fneur.2021.679927] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2021] [Accepted: 05/14/2021] [Indexed: 01/19/2023] Open
Abstract
Genetics has driven significant discoveries in the field of neurodegenerative diseases (NDDs). An emerging theme in neurodegeneration warrants an urgent and comprehensive update: that carrier status of early-onset autosomal recessive (AR) disease, typically considered benign, is associated with an increased risk of a spectrum of late-onset NDDs. Glucosylceramidase beta (GBA1) gene mutations, responsible for the AR lysosomal storage disorder Gaucher disease, are a prominent example of this principle, having been identified as an important genetic risk factor for Parkinson disease. Genetic analyses have revealed further examples, notably GRN, TREM2, EIF2AK3, and several other LSD and mitochondria function genes. In this Review, we discuss the evidence supporting the strikingly distinct allele-dependent clinical phenotypes observed in carriers of such gene mutations and its impact on the wider field of neurodegeneration.
Collapse
Affiliation(s)
| | - Huw R. Morris
- Department of Clinical and Movement Neurosciences, University College London, Queen Square Institute of Neurology, London, United Kingdom
| |
Collapse
|
27
|
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: 6] [Impact Index Per Article: 2.0] [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
|
28
|
Hummel R, Lang M, Walderbach S, Wang Y, Tegeder I, Gölz C, Schäfer MKE. Single intracerebroventricular progranulin injection adversely affects the blood-brain barrier in experimental traumatic brain injury. J Neurochem 2021; 158:342-357. [PMID: 33899947 DOI: 10.1111/jnc.15375] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2021] [Revised: 04/17/2021] [Accepted: 04/18/2021] [Indexed: 12/23/2022]
Abstract
Progranulin (PGRN) is a neurotrophic and anti-inflammatory factor with protective effects in animal models of ischemic stroke, subarachnoid hemorrhage, and traumatic brain injury (TBI). Administration of recombinant (r) PGRN prevents exaggerated brain pathology after TBI in Grn-deficient mice, suggesting that local injection of recombinant progranulin (rPGRN) provides therapeutic benefit in the acute phase of TBI. To test this hypothesis, we subjected adult male C57Bl/6N mice to the controlled cortical impact model of TBI, administered a single dose of rPGRN intracerebroventricularly (ICV) shortly before the injury, and examined behavioral and biological effects up to 5 days post injury (dpi). The anti-inflammatory bioactivity of rPGRN was confirmed by its capability to inhibit the inflammation-induced hypertrophy of murine primary microglia and astrocytes in vitro. In C57Bl/6N mice, however, ICV administration of rPGRN failed to attenuate behavioral deficits over the 5-day observation period. (Immuno)histological gene and protein expression analyses at 5 dpi did not reveal a therapeutic benefit in terms of brain injury size, brain inflammation, glia activation, cell numbers in neurogenic niches, and neuronal damage. Instead, we observed a failure of TBI-induced mRNA upregulation of the tight junction protein occludin and increased extravasation of serum immunoglobulin G into the brain parenchyma at 5 dpi. In conclusion, single ICV administration of rPGRN had not the expected protective effects in the acute phase of murine TBI, but appeared to cause an aggravation of blood-brain barrier disruption. The data raise questions about putative PGRN-boosting approaches in other types of brain injuries and disease.
Collapse
Affiliation(s)
- Regina Hummel
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Manuel Lang
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Simona Walderbach
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Yong Wang
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Irmgard Tegeder
- Institute of Clinical Pharmacology, Medical Faculty, Goethe-University Frankfurt, Frankfurt, Germany
| | - Christina Gölz
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany
| | - Michael K E Schäfer
- Department of Anesthesiology, University Medical Center of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Focus Program Translational Neurosciences (FTN) of the Johannes Gutenberg-University Mainz, Mainz, Germany.,Research Center for Immunotherapy (FZI) of the Johannes Gutenberg-University Mainz, Mainz, Germany
| |
Collapse
|
29
|
Human progranulin-expressing mice as a novel tool for the development of progranulin-modulating therapeutics. Neurobiol Dis 2021; 153:105314. [PMID: 33636385 DOI: 10.1016/j.nbd.2021.105314] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2020] [Revised: 01/24/2021] [Accepted: 02/22/2021] [Indexed: 11/24/2022] Open
Abstract
The granulin protein (also known as, and hereafter referred to as, progranulin) is a secreted glycoprotein that contributes to overall brain health. Heterozygous loss-of-function mutations in the gene encoding the progranulin protein (Granulin Precursor, GRN) are a common cause of familial frontotemporal dementia (FTD). Gene therapy approaches that aim to increase progranulin expression from a single wild-type allele, an area of active investigation for the potential treatment of GRN-dependent FTD, will benefit from the availability of a mouse model that expresses a genomic copy of the human GRN gene. Here we report the development and characterization of a novel mouse model that expresses the entire human GRN gene in its native genomic context as a single copy inserted into a defined locus (Hprt) in the mouse genome. We show that human and mouse progranulin are expressed in a similar tissue-specific pattern, suggesting that the two genes are regulated by similar mechanisms. Human progranulin rescues a phenotype characteristic of progranulin-null mice, the exaggerated and early deposition of the aging pigment lipofuscin in the brain, indicating that the two proteins are functionally similar. Longitudinal behavioural and neuropathological analyses revealed no significant differences between wild-type and human progranulin-overexpressing mice up to 18 months of age, providing evidence that long-term increase of progranulin levels is well tolerated in mice. Finally, we demonstrate that human progranulin expression can be increased in the brain using an antisense oligonucleotide that inhibits a known GRN-regulating micro-RNA, demonstrating that the transgene is responsive to potential gene therapy drugs. Human progranulin-expressing mice represent a novel and valuable tool to expedite the development of progranulin-modulating therapeutics.
Collapse
|
30
|
Abstract
Neuronal ceroid lipofuscinosis (NCLs) is a group of inherited neurodegenerative lysosomal storage diseases that together represent the most common cause of dementia in children. Phenotypically, patients have visual impairment, cognitive and motor decline, epilepsy, and premature death. A primary challenge is to halt and/or reverse these diseases, towards which developments in potential effective therapies are encouraging. Many treatments, including enzyme replacement therapy (for CLN1 and CLN2 diseases), stem-cell therapy (for CLN1, CLN2, and CLN8 diseases), gene therapy vector (for CLN1, CLN2, CLN3, CLN5, CLN6, CLN7, CLN10, and CLN11 diseases), and pharmacological drugs (for CLN1, CLN2, CLN3, and CLN6 diseases) have been evaluated for safety and efficacy in pre-clinical and clinical studies. Currently, cerliponase alpha for CLN2 disease is the only approved therapy for NCL. Lacking is any study of potential treatments for CLN4, CLN9, CLN12, CLN13 or CLN14 diseases. This review provides an overview of genetics for each CLN disease, and we discuss the current understanding from pre-clinical and clinical study of potential therapeutics. Various therapeutic interventions have been studied in many experimental animal models. Combination of treatments may be useful to slow or even halt disease progression; however, few therapies are unlikely to even partially reverse the disease and a complete reversal is currently improbable. Early diagnosis to allow initiation of therapy, when indicated, during asymptomatic stages is more important than ever.
Collapse
|
31
|
Abstract
Therapeutic viral gene delivery is an emerging technology which aims to correct genetic mutations by introducing new genetic information to cells either to correct a faulty gene or to initiate cell death in oncolytic treatments. In recent years, significant scientific progress has led to several clinical trials resulting in the approval of gene therapies for human treatment. However, successful therapies remain limited due to a number of challenges such as inefficient cell uptake, low transduction efficiency (TE), limited tropism, liver toxicity and immune response. To adress these issues and increase the number of available therapies, additives from a broad range of materials like polymers, peptides, lipids, nanoparticles, and small molecules have been applied so far. The scope of this review is to highlight these selected delivery systems from a materials perspective.
Collapse
Affiliation(s)
- Kübra Kaygisiz
- Max Planck Institute for Polymer Research, Ackermannweg 10, 55128 Mainz, Germany.
| | | |
Collapse
|
32
|
Hinderer C, Miller R, Dyer C, Johansson J, Bell P, Buza E, Wilson JM. Adeno-associated virus serotype 1-based gene therapy for FTD caused by GRN mutations. Ann Clin Transl Neurol 2020; 7:1843-1853. [PMID: 32937039 PMCID: PMC7545603 DOI: 10.1002/acn3.51165] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 06/29/2020] [Accepted: 07/31/2020] [Indexed: 11/30/2022] Open
Abstract
Objective Dominant loss‐of‐function mutations in the gene encoding the lysosomal protein, progranulin, cause 5‐10% of frontotemporal dementia cases. As progranulin undergoes secretion and endocytosis, a small number of progranulin‐expressing cells can potentially supply the protein to the entire central nervous system. Thus, gene therapy is a promising treatment approach. Methods We evaluated adeno‐associated viral vector administration into the cerebrospinal fluid as a minimally invasive approach to deliver the granulin gene to the central nervous system in a murine disease model and nonhuman primates. Results In progranulin‐deficient mice, vector delivery into the lateral cerebral ventricles increased progranulin levels in the cerebrospinal fluid and normalized histological and biochemical markers of progranulin deficiency. A single vector injection into the cisterna magna of nonhuman primates achieved CSF progranulin concentrations up to 40‐fold higher than those of normal human subjects and exceeded CSF progranulin levels of successfully treated mice. Animals treated with an adeno‐associated virus serotype 1 vector exhibited progranulin expression fivefold higher than those treated with an AAV5 vector or the AAV9 variant, AAVhu68, apparently due to remarkably efficient transduction of ependymal cells. Progranulin expression mediated by adeno‐associated viral vectors was well tolerated in nonhuman primates with no evidence of dose‐limiting toxicity, even at vector doses that induced supraphysiologic progranulin expression. Interpretation These findings support the development of AAV1‐based gene therapy for frontotemporal dementia caused by progranulin deficiency.
Collapse
Affiliation(s)
- Christian Hinderer
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Rod Miller
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Cecilia Dyer
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Julia Johansson
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Peter Bell
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - Elizabeth Buza
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| | - James M Wilson
- Gene Therapy Program, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA
| |
Collapse
|
33
|
Chaplot K, Jarvela TS, Lindberg I. Secreted Chaperones in Neurodegeneration. Front Aging Neurosci 2020; 12:268. [PMID: 33192447 PMCID: PMC7481362 DOI: 10.3389/fnagi.2020.00268] [Citation(s) in RCA: 29] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022] Open
Abstract
Protein homeostasis, or proteostasis, is a combination of cellular processes that govern protein quality control, namely, protein translation, folding, processing, and degradation. Disruptions in these processes can lead to protein misfolding and aggregation. Proteostatic disruption can lead to cellular changes such as endoplasmic reticulum or oxidative stress; organelle dysfunction; and, if continued, to cell death. A majority of neurodegenerative diseases involve the pathologic aggregation of proteins that subverts normal neuronal function. While prior reviews of neuronal proteostasis in neurodegenerative processes have focused on cytoplasmic chaperones, there is increasing evidence that chaperones secreted both by neurons and other brain cells in the extracellular - including transsynaptic - space play important roles in neuronal proteostasis. In this review, we will introduce various secreted chaperones involved in neurodegeneration. We begin with clusterin and discuss its identification in various protein aggregates, and the use of increased cerebrospinal fluid (CSF) clusterin as a potential biomarker and as a potential therapeutic. Our next secreted chaperone is progranulin; polymorphisms in this gene represent a known genetic risk factor for frontotemporal lobar degeneration, and progranulin overexpression has been found to be effective in reducing Alzheimer's- and Parkinson's-like neurodegenerative phenotypes in mouse models. We move on to BRICHOS domain-containing proteins, a family of proteins containing highly potent anti-amyloidogenic activity; we summarize studies describing the biochemical mechanisms by which recombinant BRICHOS protein might serve as a therapeutic agent. The next section of the review is devoted to the secreted chaperones 7B2 and proSAAS, small neuronal proteins which are packaged together with neuropeptides and released during synaptic activity. Since proteins can be secreted by both classical secretory and non-classical mechanisms, we also review the small heat shock proteins (sHsps) that can be secreted from the cytoplasm to the extracellular environment and provide evidence for their involvement in extracellular proteostasis and neuroprotection. Our goal in this review focusing on extracellular chaperones in neurodegenerative disease is to summarize the most recent literature relating to neurodegeneration for each secreted chaperone; to identify any common mechanisms; and to point out areas of similarity as well as differences between the secreted chaperones identified to date.
Collapse
Affiliation(s)
| | | | - Iris Lindberg
- Department of Anatomy and Neurobiology, University of Maryland School of Medicine, University of Maryland, Baltimore, Baltimore, MD, United States
| |
Collapse
|
34
|
Telpoukhovskaia MA, Liu K, Sayed FA, Etchegaray JI, Xie M, Zhan L, Li Y, Zhou Y, Le D, Bahr BA, Bogyo M, Ding S, Gan L. Discovery of small molecules that normalize the transcriptome and enhance cysteine cathepsin activity in progranulin-deficient microglia. Sci Rep 2020; 10:13688. [PMID: 32792571 PMCID: PMC7426857 DOI: 10.1038/s41598-020-70534-9] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2020] [Accepted: 07/17/2020] [Indexed: 12/11/2022] Open
Abstract
Patients with frontotemporal dementia (FTD) resulting from granulin (GRN) haploinsufficiency have reduced levels of progranulin and exhibit dysregulation in inflammatory and lysosomal networks. Microglia produce high levels of progranulin, and reduction of progranulin in microglia alone is sufficient to recapitulate inflammation, lysosomal dysfunction, and hyperproliferation in a cell-autonomous manner. Therefore, targeting microglial dysfunction caused by progranulin insufficiency represents a potential therapeutic strategy to manage neurodegeneration in FTD. Limitations of current progranulin-enhancing strategies necessitate the discovery of new targets. To identify compounds that can reverse microglial defects in Grn-deficient mouse microglia, we performed a compound screen coupled with high throughput sequencing to assess key transcriptional changes in inflammatory and lysosomal pathways. Positive hits from this initial screen were then further narrowed down based on their ability to rescue cathepsin activity, a critical biochemical readout of lysosomal capacity. The screen identified nor-binaltorphimine dihydrochloride (nor-BNI) and dibutyryl-cAMP, sodium salt (DB-cAMP) as two phenotypic modulators of progranulin deficiency. In addition, nor-BNI and DB-cAMP also rescued cell cycle abnormalities in progranulin-deficient cells. These data highlight the potential of a transcription-based platform for drug screening, and advance two novel lead compounds for FTD.
Collapse
Affiliation(s)
- Maria A Telpoukhovskaia
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA.,Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Kai Liu
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, 94158, USA
| | - Faten A Sayed
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA.,Department of Neurology, University of California, San Francisco, CA, 94158, USA.,Neuroscience Graduate Program, University of California, San Francisco, CA, 94158, USA
| | | | - Min Xie
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA.,Gladstone Institute of Cardiovascular Disease, San Francisco, CA, 94158, USA
| | - Lihong Zhan
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA.,Department of Neurology, University of California, San Francisco, CA, 94158, USA
| | - Yaqiao Li
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
| | - Yungui Zhou
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
| | - David Le
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA
| | - Ben A Bahr
- Biotechnology Research and Training Center, University of North Carolina At Pembroke, Pembroke, NC, 28372, USA
| | - Matthew Bogyo
- Department of Pathology, Stanford University, Stanford, CA, 94305, USA
| | - Sheng Ding
- Gladstone Institute of Cardiovascular Disease, San Francisco, CA, 94158, USA
| | - Li Gan
- Gladstone Institute of Neurological Disease, San Francisco, CA, 94158, USA. .,Department of Neurology, University of California, San Francisco, CA, 94158, USA. .,Helen and Robert Appel Alzheimer's Disease Research Institute, Brain and Mind Research Institute, Weill Cornell Medicine, New York, NY, 10021, USA.
| |
Collapse
|
35
|
Mijanović O, Branković A, Borovjagin AV, Butnaru DV, Bezrukov EA, Sukhanov RB, Shpichka A, Timashev P, Ulasov I. Battling Neurodegenerative Diseases with Adeno-Associated Virus-Based Approaches. Viruses 2020; 12:E460. [PMID: 32325732 PMCID: PMC7232215 DOI: 10.3390/v12040460] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2020] [Revised: 04/06/2020] [Accepted: 04/15/2020] [Indexed: 02/06/2023] Open
Abstract
Neurodegenerative diseases (NDDs) are most commonly found in adults and remain essentially incurable. Gene therapy using AAV vectors is a rapidly-growing field of experimental medicine that holds promise for the treatment of NDDs. To date, the delivery of a therapeutic gene into target cells via AAV represents a major obstacle in the field. Ideally, transgenes should be delivered into the target cells specifically and efficiently, while promiscuous or off-target gene delivery should be minimized to avoid toxicity. In the pursuit of an ideal vehicle for NDD gene therapy, a broad variety of vector systems have been explored. Here we specifically outline the advantages of adeno-associated virus (AAV)-based vector systems for NDD therapy application. In contrast to many reviews on NDDs that can be found in the literature, this review is rather focused on AAV vector selection and their preclinical testing in experimental and preclinical NDD models. Preclinical and in vitro data reveal the strong potential of AAV for NDD-related diagnostics and therapeutic strategies.
Collapse
Affiliation(s)
- Olja Mijanović
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia;
| | - Ana Branković
- Department of Forensics, University of Criminal Investigation and Police Studies, Belgrade 11000, Serbia;
| | - Anton V. Borovjagin
- Department of Biomedical Engineering, University of Alabama at Birmingham, Birmingham, AL 35294, USA;
| | - Denis V. Butnaru
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.V.B.); (A.S.); (P.T.)
| | - Evgeny A. Bezrukov
- Institute for Uronephrology and Reproductive Health, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (E.A.B.); (R.B.S.)
| | - Roman B. Sukhanov
- Institute for Uronephrology and Reproductive Health, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (E.A.B.); (R.B.S.)
| | - Anastasia Shpichka
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.V.B.); (A.S.); (P.T.)
| | - Peter Timashev
- Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia; (D.V.B.); (A.S.); (P.T.)
- Institute of Photonic Technologies, Research Center “Crystallography and Photonics”, Russian Academy of Sciences, Troitsk, Moscow 142190, Russia
- Department of Polymers and Composites, N.N. Semenov Institute of Chemical Physics, Moscow 119991, Russia
- Chemistry Department, Lomonosov Moscow State University, Moscow 119991, Russia
| | - Ilya Ulasov
- Group of Experimental Biotherapy and Diagnostics, Institute for Regenerative Medicine, Sechenov First Moscow State Medical University, Moscow 119991, Russia;
| |
Collapse
|
36
|
Liu W, Kleine-Holthaus SM, Herranz-Martin S, Aristorena M, Mole SE, Smith AJ, Ali RR, Rahim AA. Experimental gene therapies for the NCLs. Biochim Biophys Acta Mol Basis Dis 2020; 1866:165772. [PMID: 32220628 DOI: 10.1016/j.bbadis.2020.165772] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2019] [Revised: 03/16/2020] [Accepted: 03/17/2020] [Indexed: 02/06/2023]
Abstract
The neuronal ceroid lipofuscinoses (NCLs), also known as Batten disease, are a group of rare monogenic neurodegenerative diseases predominantly affecting children. All NCLs are lethal and incurable and only one has an approved treatment available. To date, 13 NCL subtypes (CLN1-8, CLN10-14) have been identified, based on the particular disease-causing defective gene. The exact functions of NCL proteins and the pathological mechanisms underlying the diseases are still unclear. However, gene therapy has emerged as an attractive therapeutic strategy for this group of conditions. Here we provide a short review discussing updates on the current gene therapy studies for the NCLs.
Collapse
Affiliation(s)
- Wenfei Liu
- UCL School of Pharmacy, University College London, UK
| | | | - Saul Herranz-Martin
- UCL School of Pharmacy, University College London, UK; Centro de Biología Molecular Severo Ochoa (UAM-CSIC) and Departamento de Biología Molecular,Universidad Autónoma de Madrid (UAM), Madrid, Spain
| | | | - Sara E Mole
- MRC Laboratory for Molecular Cell Biology, University College London, Gower Street, London WC1E 6BT, UK; UCL Great Ormond Street Institute of Child Health, 30 Guildford Street, London WC1N 1EH, UK
| | | | - Robin R Ali
- UCL Institute of Ophthalmology, University College London, UK; NIHR Biomedical Research Centre at Moorfields Eye Hospital NHS Foundation Trust, UK
| | - Ahad A Rahim
- UCL School of Pharmacy, University College London, UK.
| |
Collapse
|
37
|
Abstract
Several viral vector-based gene therapy drugs have now received marketing approval. A much larger number of additional viral vectors are in various stages of clinical trials for the treatment of genetic and acquired diseases, with many more in pre-clinical testing. Efficiency of gene transfer and ability to provide long-term therapy make these vector systems very attractive. In fact, viral vector gene therapy has been able to treat or even cure diseases for which there had been no or only suboptimal treatments. However, innate and adaptive immune responses to these vectors and their transgene products constitute substantial hurdles to clinical development and wider use in patients. This review provides an overview of the type of immune responses that have been documented in animal models and in humans who received gene transfer with one of three widely tested vector systems, namely adenoviral, lentiviral, or adeno-associated viral vectors. Particular emphasis is given to mechanisms leading to immune responses, efforts to reduce vector immunogenicity, and potential solutions to the problems. At the same time, we point out gaps in our knowledge that should to be filled and problems that need to be addressed going forward.
Collapse
Affiliation(s)
- Jamie L Shirley
- Gene Therapy Center, University of Massachusetts, Worchester, MA, USA
| | - Ype P de Jong
- Division of Gastroenterology and Hepatology, Weill Cornell Medicine, New York, NY, USA
| | - Cox Terhorst
- Division of Immunology, Beth Israel Deaconess Medical Center (BIDMC), Harvard Medical School, Boston, MA, USA
| | - Roland W Herzog
- Herman B Wells Center for Pediatric Research, Indiana University School of Medicine, Indianapolis, IN, USA.
| |
Collapse
|
38
|
Rosenberg JB, Chen A, Kaminsky SM, Crystal RG, Sondhi D. Advances in the Treatment of Neuronal Ceroid Lipofuscinosis. Expert Opin Orphan Drugs 2019; 7:473-500. [PMID: 33365208 PMCID: PMC7755158 DOI: 10.1080/21678707.2019.1684258] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/21/2019] [Accepted: 10/21/2019] [Indexed: 12/27/2022]
Abstract
Neuronal ceroid lipofuscinoses (NCL) represent a class of neurodegenerative disorders involving defective lysosomal processing enzymes or receptors, leading to lysosomal storage disorders, typically characterized by observation of cognitive and visual impairments, epileptic seizures, ataxia, and deterioration of motor skills. Recent success of a biologic (Brineura®) for the treatment of neurologic manifestations of the central nervous system (CNS) has led to renewed interest in therapeutics for NCL, with the goal of ablating or reversing the impact of these devastating disorders. Despite complex challenges associated with CNS therapy, many treatment modalities have been evaluated, including enzyme replacement therapy, gene therapy, stem cell therapy, and small molecule pharmacotherapy. Because the clinical endpoints for the evaluation of candidate therapies are complex and often reliant on subjective clinical scales, the development of quantitative biomarkers for NCLs has become an apparent necessity for the validation of potential treatments. We will discuss the latest findings in the search for relevant biomarkers for assessing disease progression. For this review, we will focus primarily on recent pre-clinical and clinical developments for treatments to halt or cure these NCL diseases. Continued development of current therapies and discovery of newer modalities will be essential for successful therapeutics for NCL. AREAS COVERED The reader will be introduced to the NCL subtypes, natural histories, experimental animal models, and biomarkers for NCL progression; challenges and different therapeutic approaches, and the latest pre-clinical and clinical research for therapeutic development for the various NCLs. This review corresponds to the literatures covering the years from 1968 to mid-2019, but primarily addresses pre-clinical and clinical developments for the treatment of NCL disease in the last decade and as a follow-up to our 2013 review of the same topic in this journal. EXPERT OPINION Much progress has been made in the treatment of neurologic diseases, such as the NCLs, including better animal models and improved therapeutics with better survival outcomes. Encouraging results are being reported at symposiums and in the literature, with multiple therapeutics reaching the clinical trial stage for the NCLs. The potential for a cure could be at hand after many years of trial and error in the preclinical studies. The clinical development of enzyme replacement therapy (Brineura® for CLN2), immunosuppression (CellCept® for CLN3), and gene therapy vectors (for CLN1, CLN2, CLN3, and CLN6) are providing encouragement to families that have a child afflicted with NCL. We believe that successful therapies in the future may involve the combination of two or more therapeutic modalities to provide therapeutic benefit especially as the patients grow older.
Collapse
Affiliation(s)
- Jonathan B Rosenberg
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Alvin Chen
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Stephen M Kaminsky
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Ronald G Crystal
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| | - Dolan Sondhi
- Department of Genetic Medicine, Weill Cornell Medical College, New York, New York
| |
Collapse
|
39
|
Huber RJ, Hughes SM, Liu W, Morgan A, Tuxworth RI, Russell C. The contribution of multicellular model organisms to neuronal ceroid lipofuscinosis research. Biochim Biophys Acta Mol Basis Dis 2019; 1866:165614. [PMID: 31783156 DOI: 10.1016/j.bbadis.2019.165614] [Citation(s) in RCA: 21] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 11/14/2019] [Accepted: 11/15/2019] [Indexed: 02/07/2023]
Abstract
The NCLs (neuronal ceroid lipofuscinosis) are forms of neurodegenerative disease that affect people of all ages and ethnicities but are most prevalent in children. Commonly known as Batten disease, this debilitating neurological disorder is comprised of 13 different subtypes that are categorized based on the particular gene that is mutated (CLN1-8, CLN10-14). The pathological mechanisms underlying the NCLs are not well understood due to our poor understanding of the functions of NCL proteins. Only one specific treatment (enzyme replacement therapy) is approved, which is for the treating the brain in CLN2 disease. Hence there remains a desperate need for further research into disease-modifying treatments. In this review, we present and evaluate the genes, proteins and studies performed in the social amoeba, nematode, fruit fly, zebrafish, mouse and large animals pertinent to NCL. In particular, we highlight the use of multicellular model organisms to study NCL protein function, pathology and pathomechanisms. Their use in testing novel therapeutic approaches is also presented. With this information, we highlight how future research in these systems may be able to provide new insight into NCL protein functions in human cells and aid in the development of new therapies.
Collapse
Affiliation(s)
- Robert J Huber
- Department of Biology, Trent University, Peterborough, Ontario K9L 0G2, Canada
| | - Stephanie M Hughes
- Department of Biochemistry, School of Biomedical Sciences, Brain Health Research Centre and Genetics Otago, University of Otago, Dunedin, New Zealand
| | - Wenfei Liu
- School of Pharmacy, University College London, London, WC1N 1AX, UK
| | - Alan Morgan
- Department of Cellular and Molecular Physiology, Institute of Translational Medicine, University of Liverpool, Crown St., Liverpool L69 3BX, UK
| | - Richard I Tuxworth
- Institute of Cancer and Genomic Sciences, University of Birmingham, Birmingham B15 2TT, UK
| | - Claire Russell
- Dept. Comparative Biomedical Sciences, Royal Veterinary College, Royal College Street, London NW1 0TU, UK.
| |
Collapse
|
40
|
Butler VJ, Gao F, Corrales CI, Cortopassi WA, Caballero B, Vohra M, Ashrafi K, Cuervo AM, Jacobson MP, Coppola G, Kao AW. Age- and stress-associated C. elegans granulins impair lysosomal function and induce a compensatory HLH-30/TFEB transcriptional response. PLoS Genet 2019; 15:e1008295. [PMID: 31398187 PMCID: PMC6703691 DOI: 10.1371/journal.pgen.1008295] [Citation(s) in RCA: 20] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/22/2019] [Revised: 08/21/2019] [Accepted: 07/07/2019] [Indexed: 12/11/2022] Open
Abstract
The progressive failure of protein homeostasis is a hallmark of aging and a common feature in neurodegenerative disease. As the enzymes executing the final stages of autophagy, lysosomal proteases are key contributors to the maintenance of protein homeostasis with age. We previously reported that expression of granulin peptides, the cleavage products of the neurodegenerative disease protein progranulin, enhance the accumulation and toxicity of TAR DNA binding protein 43 (TDP-43) in Caenorhabditis elegans (C. elegans). In this study we show that C. elegans granulins are produced in an age- and stress-dependent manner. Granulins localize to the endolysosomal compartment where they impair lysosomal protease expression and activity. Consequently, protein homeostasis is disrupted, promoting the nuclear translocation of the lysosomal transcription factor HLH-30/TFEB, and prompting cells to activate a compensatory transcriptional program. The three C. elegans granulin peptides exhibited distinct but overlapping functional effects in our assays, which may be due to amino acid composition that results in distinct electrostatic and hydrophobicity profiles. Our results support a model in which granulin production modulates a critical transition between the normal, physiological regulation of protease activity and the impairment of lysosomal function that can occur with age and disease.
Collapse
Affiliation(s)
- Victoria J. Butler
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California, United States of America
| | - Fuying Gao
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Christian I. Corrales
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California, United States of America
| | - Wilian A. Cortopassi
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California, United States of America
| | - Benjamin Caballero
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, New York, United States of America
| | - Mihir Vohra
- Department of Physiology, University of California, San Francisco, California, United States of America
| | - Kaveh Ashrafi
- Department of Physiology, University of California, San Francisco, California, United States of America
| | - Ana Maria Cuervo
- Department of Developmental and Molecular Biology, Albert Einstein College of Medicine, New York, New York, United States of America
| | - Matthew P. Jacobson
- Department of Pharmaceutical Chemistry, University of California, San Francisco, California, United States of America
| | - Giovanni Coppola
- Semel Institute for Neuroscience and Human Behavior, Departments of Psychiatry and Biobehavioral Sciences and Neurology, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California, United States of America
| | - Aimee W. Kao
- Memory and Aging Center, Department of Neurology, University of California, San Francisco, California, United States of America
- * E-mail:
| |
Collapse
|
41
|
An update on genetic frontotemporal dementia. J Neurol 2019; 266:2075-2086. [PMID: 31119452 PMCID: PMC6647117 DOI: 10.1007/s00415-019-09363-4] [Citation(s) in RCA: 223] [Impact Index Per Article: 44.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Revised: 05/01/2019] [Accepted: 05/03/2019] [Indexed: 12/12/2022]
Abstract
Frontotemporal dementia (FTD) is a highly heritable group of neurodegenerative disorders, with around 30% of patients having a strong family history. The majority of that heritability is accounted for by autosomal dominant mutations in the chromosome 9 open reading frame 72 (C9orf72), progranulin (GRN), and microtubule-associated protein tau (MAPT) genes, with mutations more rarely seen in a number of other genes. This review will discuss the recent updates in the field of genetic FTD. Age at symptom onset in genetic FTD is variable with recently identified genetic modifiers including TMEM106B (in GRN carriers particularly) and a polymorphism at a locus containing two overlapping genes LOC101929163 and C6orf10 (in C9orf72 carriers). Behavioural variant FTD (bvFTD) is the most common diagnosis in each of the genetic groups, although in C9orf72 carriers amyotrophic lateral sclerosis either alone, or with bvFTD, is also common. An atypical neuropsychiatric presentation is also seen in C9orf72 carriers and family members of carriers are at greater risk of psychiatric disorders including schizophrenia and autistic spectrum disorders. Large natural history studies of presymptomatic genetic FTD are now underway both in Europe/Canada (GENFI—the Genetic FTD Initiative) and in the US (ARTFL/LEFFTDS study), collaborating together under the banner of the FTD Prevention Initiative (FPI). These studies are taking forward the validation of cognitive, imaging and fluid biomarkers that aim to robustly measure disease onset, staging and progression in genetic FTD. Grey matter changes on MRI and hypometabolism on FDG-PET are seen at least 10 years before symptom onset with white matter abnormalities seen earlier, but the pattern and exact timing of changes differ between different genetic groups. In contrast, tau PET has yet to show promise in genetic FTD. Three key fluid biomarkers have been identified so far that are likely to be helpful in clinical trials—CSF or blood neurofilament light chain levels (in all groups), CSF or blood progranulin levels (in GRN carriers) and CSF poly(GP) dipeptide repeat protein levels (in C9orf72 carriers). Increased knowledge about genetic FTD has led to more clinical presymptomatic genetic testing but this has not yet been mirrored in the development of either an accepted FTD-specific testing protocol or provision of appropriate psychological support mechanisms for those living through the at-risk phase. This will become even more relevant as disease-modifying therapy trials start in each of the genetic groups over the next few years.
Collapse
|
42
|
Microglial Progranulin: Involvement in Alzheimer's Disease and Neurodegenerative Diseases. Cells 2019; 8:cells8030230. [PMID: 30862089 PMCID: PMC6468562 DOI: 10.3390/cells8030230] [Citation(s) in RCA: 48] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/22/2018] [Revised: 03/06/2019] [Accepted: 03/07/2019] [Indexed: 12/12/2022] Open
Abstract
Neurodegenerative diseases such as Alzheimer’s disease have proven resistant to new treatments. The complexity of neurodegenerative disease mechanisms can be highlighted by accumulating evidence for a role for a growth factor, progranulin (PGRN). PGRN is a glycoprotein encoded by the GRN/Grn gene with multiple cellular functions, including neurotrophic, anti-inflammatory and lysosome regulatory properties. Mutations in the GRN gene can lead to frontotemporal lobar degeneration (FTLD), a cause of dementia, and neuronal ceroid lipofuscinosis (NCL), a lysosomal storage disease. Both diseases are associated with loss of PGRN function resulting, amongst other features, in enhanced microglial neuroinflammation and lysosomal dysfunction. PGRN has also been implicated in Alzheimer’s disease (AD). Unlike FTLD, increased expression of PGRN occurs in brains of human AD cases and AD model mice, particularly in activated microglia. How microglial PGRN might be involved in AD and other neurodegenerative diseases will be discussed. A unifying feature of PGRN in diseases might be its modulation of lysosomal function in neurons and microglia. Many experimental models have focused on consequences of PGRN gene deletion: however, possible outcomes of increasing PGRN on microglial inflammation and neurodegeneration will be discussed. We will also suggest directions for future studies on PGRN and microglia in relation to neurodegenerative diseases.
Collapse
|
43
|
Cui Y, Hettinghouse A, Liu CJ. Progranulin: A conductor of receptors orchestra, a chaperone of lysosomal enzymes and a therapeutic target for multiple diseases. Cytokine Growth Factor Rev 2019; 45:53-64. [PMID: 30733059 DOI: 10.1016/j.cytogfr.2019.01.002] [Citation(s) in RCA: 55] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2019] [Accepted: 01/29/2019] [Indexed: 12/14/2022]
Abstract
Progranulin (PGRN), a widely expressed glycoprotein with pleiotropic function, has been linked to a host of physiological processes and diverse pathological states. A series of contemporary preclinical disease models and clinical trials have evaluated various therapeutic strategies targeting PGRN, highlighting PGRN as a promising therapeutic target. Herein we summarize available knowledge of PGRN targeting in various kinds of diseases, including common neurological diseases, inflammatory autoimmune diseases, cancer, tissue repair, and rare lysosomal storage diseases, with a focus on the functional domain-oriented drug development strategies. In particular, we emphasize the role of extracellular PGRN as a non-conventional, extracellular matrix bound, growth factor-like conductor orchestrating multiple membrane receptors and intracellular PGRN as a chaperone/co-chaperone that mediates the folding and traffic of its various binding partners.
Collapse
Affiliation(s)
- Yazhou Cui
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003, USA; Shandong Medical Biotechnological Center, Shandong Academy of Medical Sciences, Jinan, 250062, China
| | - Aubryanna Hettinghouse
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003, USA
| | - Chuan-Ju Liu
- Department of Orthopaedic Surgery, New York University Medical Center, New York, NY, 10003, USA; Department of Cell Biology, New York University School of Medicine, New York, NY, 10016, USA.
| |
Collapse
|