1
|
Sleat DE, Banach-Petrosky W, Larrimore KE, Nemtsova Y, Wiseman JA, Najafi A, Johnson D, Poole TA, Takahashi K, Cooper JD, Lobel P. A mouse mutant deficient in both neuronal ceroid lipofuscinosis-associated proteins CLN3 and TPP1. J Inherit Metab Dis 2023; 46:720-734. [PMID: 37078466 PMCID: PMC10330656 DOI: 10.1002/jimd.12619] [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: 01/25/2023] [Revised: 04/11/2023] [Accepted: 04/17/2023] [Indexed: 04/21/2023]
Abstract
Late-infantile neuronal ceroid lipofuscinosis (LINCL) and juvenile neuronal ceroid lipofuscinosis (JNCL) are inherited neurodegenerative diseases caused by mutations in the genes encoding lysosomal proteins tripeptidyl peptidase 1 (TPP1) and CLN3 protein, respectively. TPP1 is well-understood and, aided by animal models that accurately recapitulate the human disease, enzyme replacement therapy has been approved and other promising therapies are emerging. In contrast, there are no effective treatments for JNCL, partly because the function of the CLN3 protein remains unknown but also because animal models have attenuated disease and lack robust survival phenotypes. Mouse models for LINCL and JNCL, with mutations in Tpp1 and Cln3, respectively, have been thoroughly characterized but the phenotype of a double Cln3/Tpp1 mutant remains unknown. We created this double mutant and find that its phenotype is essentially indistinguishable from the single Tpp1-/- mutant in terms of survival and brain pathology. Analysis of brain proteomic changes in the single Tpp1-/- and double Cln3-/- ;Tpp1-/- mutants indicates largely overlapping sets of altered proteins and reinforces earlier studies that highlight GPNMB, LYZ2, and SERPINA3 as promising biomarker candidates in LINCL while several lysosomal proteins including SMPD1 and NPC1 appear to be altered in the Cln3-/- animals. An unexpected finding was that Tpp1 heterozygosity significantly decreased lifespan of the Cln3-/- mouse. The truncated survival of this mouse model makes it potentially useful in developing therapies for JNCL using survival as an endpoint. In addition, this model may also provide insights into CLN3 protein function and its potential functional interactions with TPP1.
Collapse
Affiliation(s)
- David E. Sleat
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers Biomedical Health Sciences, Rutgers University, Piscataway, NJ, United States of America
| | - Whitney Banach-Petrosky
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
| | - Katherine E. Larrimore
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
| | - Yuliya Nemtsova
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
| | - Jennifer A. Wiseman
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
| | - Allison Najafi
- The Lundquist Institute (formerly Los Angeles Biomedical Research Institute), Harbor-UCLA Medical Center, and David Geffen School of Medicine, University of California, Los Angeles, Torrance, CA United States of America
| | - Dymonn Johnson
- Departments of Pediatrics, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
| | - Timothy A. Poole
- Departments of Pediatrics, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
| | - Keigo Takahashi
- Departments of Pediatrics, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
| | - Jonathan D. Cooper
- The Lundquist Institute (formerly Los Angeles Biomedical Research Institute), Harbor-UCLA Medical Center, and David Geffen School of Medicine, University of California, Los Angeles, Torrance, CA United States of America
- Departments of Pediatrics, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
- Genetics, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
- Neurology, Division of Genetics and Genomic Medicine, Washington University in St Louis, School of Medicine, St Louis, MO, United States of America
| | - Peter Lobel
- Center for Advanced Biotechnology and Medicine, Rutgers University, Piscataway, NJ, United States of America
- Department of Biochemistry and Molecular Biology, Robert Wood Johnson Medical School, Rutgers Biomedical Health Sciences, Rutgers University, Piscataway, NJ, United States of America
| |
Collapse
|
2
|
Nittari G, Tomassoni D, Roy P, Martinelli I, Tayebati SK, Amenta F. Batten disease through different in vivo and in vitro models: A review. J Neurosci Res 2023; 101:298-315. [PMID: 36434776 DOI: 10.1002/jnr.25147] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Revised: 10/25/2022] [Accepted: 11/09/2022] [Indexed: 11/27/2022]
Abstract
Batten disease consists of a family of primarily autosomal recessive, progressive neuropediatric disorders, also known as neuronal ceroid lipofuscinoses (NCLs). These pathologies are characterized by seizures and visual, cognitive and motor decline, and premature death. The pathophysiology of this rare disease is still unclear despite the years of trials and financial aids. This paper has reviewed advantages and limits of in vivo and in vitro models of Batten disease from murine and larger animal models to primitive unicellular models, until the most recently developed patient-derived induced pluripotent stem cells. For each model advantages, limits and applications were analyzed. The first prototypes investigated were murine models that due to their limits were replaced by larger animals. In vitro models gradually replaced animal models for practical, cost, and ethical reasons. Using induced pluripotent stem cells to study neurodegeneration is a new way of studying the disease, since they can be distinguished into differentiating elements like neurons, which are susceptible to neurodegeneration. In vivo and in vitro models have contributed to clarifying to some extent the pathophysiology of the disease. The collection and sharing of suitable human bio samples likely through biobanks can contribute to a better understanding, prevention, and to identify possible treatment strategies of Batten disease.
Collapse
Affiliation(s)
- Giulio Nittari
- School of Medicinal and Health Products Sciences, Clinical Research, Telemedicine and Telepharmacy Center, University of Camerino, Camerino, Italy
| | - Daniele Tomassoni
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Proshanta Roy
- School of Biosciences and Veterinary Medicine, University of Camerino, Camerino, Italy
| | - Ilenia Martinelli
- School of Medicinal and Health Products Sciences, Clinical Research, Telemedicine and Telepharmacy Center, University of Camerino, Camerino, Italy
| | - Seyed Khosrow Tayebati
- School of Medicinal and Health Products Sciences, Clinical Research, Telemedicine and Telepharmacy Center, University of Camerino, Camerino, Italy
| | - Francesco Amenta
- School of Medicinal and Health Products Sciences, Clinical Research, Telemedicine and Telepharmacy Center, University of Camerino, Camerino, Italy
| |
Collapse
|
3
|
Chen YH, Tian W, Yasuda M, Ye Z, Song M, Mandel U, Kristensen C, Povolo L, Marques ARA, Čaval T, Heck AJR, Sampaio JL, Johannes L, Tsukimura T, Desnick R, Vakhrushev SY, Yang Z, Clausen H. A universal GlycoDesign for lysosomal replacement enzymes to improve circulation time and biodistribution. Front Bioeng Biotechnol 2023; 11:1128371. [PMID: 36911201 PMCID: PMC9999025 DOI: 10.3389/fbioe.2023.1128371] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2022] [Accepted: 02/06/2023] [Indexed: 03/14/2023] Open
Abstract
Currently available enzyme replacement therapies for lysosomal storage diseases are limited in their effectiveness due in part to short circulation times and suboptimal biodistribution of the therapeutic enzymes. We previously engineered Chinese hamster ovary (CHO) cells to produce α-galactosidase A (GLA) with various N-glycan structures and demonstrated that elimination of mannose-6-phosphate (M6P) and conversion to homogeneous sialylated N-glycans prolonged circulation time and improved biodistribution of the enzyme following a single-dose infusion into Fabry mice. Here, we confirmed these findings using repeated infusions of the glycoengineered GLA into Fabry mice and further tested whether this glycoengineering approach, Long-Acting-GlycoDesign (LAGD), could be implemented on other lysosomal enzymes. LAGD-engineered CHO cells stably expressing a panel of lysosomal enzymes [aspartylglucosamine (AGA), beta-glucuronidase (GUSB), cathepsin D (CTSD), tripeptidyl peptidase (TPP1), alpha-glucosidase (GAA) or iduronate 2-sulfatase (IDS)] successfully converted all M6P-containing N-glycans to complex sialylated N-glycans. The resulting homogenous glycodesigns enabled glycoprotein profiling by native mass spectrometry. Notably, LAGD extended the plasma half-life of all three enzymes tested (GLA, GUSB, AGA) in wildtype mice. LAGD may be widely applicable to lysosomal replacement enzymes to improve their circulatory stability and therapeutic efficacy.
Collapse
Affiliation(s)
- Yen-Hsi Chen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,GlycoDisplay ApS, Copenhagen, Denmark
| | - Weihua Tian
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Department of Biotechnology and Biomedicine, Technical University of Denmark, Lyngby, Denmark
| | - Makiko Yasuda
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Zilu Ye
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk Foundation Center for Protein Research, Proteomics Program, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ming Song
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Ulla Mandel
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Lorenzo Povolo
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | | | - Tomislav Čaval
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Science4Life, Utrecht University and Netherlands Proteomics Centre, Utrecht, Netherlands
| | - Albert J R Heck
- Biomolecular Mass Spectrometry and Proteomics, Bijvoet Center for Biomolecular Research and Utrecht Institute for Pharmaceutical Sciences, Science4Life, Utrecht University and Netherlands Proteomics Centre, Utrecht, Netherlands
| | - Julio Lopes Sampaio
- Institut Curie, PSL Research University, Cellular and Chemical Biology, U1143 INSERM, UMR3666 CNRS, Paris, France
| | - Ludger Johannes
- Institut Curie, PSL Research University, Cellular and Chemical Biology, U1143 INSERM, UMR3666 CNRS, Paris, France
| | - Takahiro Tsukimura
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States.,Department of Functional Bioanalysis, Meiji Pharmaceutical University, Tokyo, Japan
| | - Robert Desnick
- Department of Genetics and Genomic Sciences, Icahn School of Medicine at Mount Sinai, New York, NY, United States
| | - Sergey Y Vakhrushev
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| | - Zhang Yang
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark.,Novo Nordisk AS, Copenhagen, Denmark
| | - Henrik Clausen
- Copenhagen Center for Glycomics, Department of Cellular and Molecular Medicine, Faculty of Health and Medical Sciences, University of Copenhagen, Copenhagen, Denmark
| |
Collapse
|
4
|
Pardridge WM. A Historical Review of Brain Drug Delivery. Pharmaceutics 2022; 14:1283. [PMID: 35745855 PMCID: PMC9229021 DOI: 10.3390/pharmaceutics14061283] [Citation(s) in RCA: 80] [Impact Index Per Article: 26.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2022] [Revised: 06/01/2022] [Accepted: 06/07/2022] [Indexed: 12/13/2022] Open
Abstract
The history of brain drug delivery is reviewed beginning with the first demonstration, in 1914, that a drug for syphilis, salvarsan, did not enter the brain, due to the presence of a blood-brain barrier (BBB). Owing to restricted transport across the BBB, FDA-approved drugs for the CNS have been generally limited to lipid-soluble small molecules. Drugs that do not cross the BBB can be re-engineered for transport on endogenous BBB carrier-mediated transport and receptor-mediated transport systems, which were identified during the 1970s-1980s. By the 1990s, a multitude of brain drug delivery technologies emerged, including trans-cranial delivery, CSF delivery, BBB disruption, lipid carriers, prodrugs, stem cells, exosomes, nanoparticles, gene therapy, and biologics. The advantages and limitations of each of these brain drug delivery technologies are critically reviewed.
Collapse
Affiliation(s)
- William M Pardridge
- Department of Medicine, University of California, Los Angeles (UCLA), Los Angeles, CA 90095, USA
| |
Collapse
|
5
|
Pardridge WM. Blood-brain barrier delivery for lysosomal storage disorders with IgG-lysosomal enzyme fusion proteins. Adv Drug Deliv Rev 2022; 184:114234. [PMID: 35307484 DOI: 10.1016/j.addr.2022.114234] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2021] [Revised: 02/25/2022] [Accepted: 03/14/2022] [Indexed: 12/12/2022]
Abstract
The majority of lysosomal storage diseases affect the brain. Treatment of the brain with intravenous enzyme replacement therapy is not successful, because the recombinant lysosomal enzymes do not cross the blood-brain barrier (BBB). Biologic drugs, including lysosomal enzymes, can be re-engineered for BBB delivery as IgG-enzyme fusion proteins. The IgG domain of the fusion protein is a monoclonal antibody directed against an endogenous receptor-mediated transporter at the BBB, such as the insulin receptor or the transferrin receptor. This receptor transports the IgG across the BBB, in parallel with the endogenous receptor ligand, and the IgG acts as a molecular Trojan horse to ferry into brain the lysosomal enzyme genetically fused to the IgG. The IgG-enzyme fusion protein is bi-functional and retains both high affinity binding for the BBB receptor, and high lysosomal enzyme activity. IgG-lysosomal enzymes are presently in clinical trials for treatment of the brain in Mucopolysaccharidosis.
Collapse
|
6
|
Singh RB, Gupta P, Kartik A, Farooqui N, Singhal S, Shergill S, Singh KP, Agarwal A. Ocular Manifestations of Neuronal Ceroid Lipofuscinoses. Semin Ophthalmol 2021; 36:582-595. [PMID: 34106804 DOI: 10.1080/08820538.2021.1936571] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Neuronal ceroid lipofuscinoses (NCLs) are a group of rare neurodegenerative storage disorders associated with devastating visual prognosis, with an incidence of 1/1,000,000 in the United States and comparatively higher incidence in European countries. The pathophysiological mechanisms causing NCLs occur due to enzymatic or transmembrane defects in various sub-cellular organelles including lysosomes, endoplasmic reticulum, and cytoplasmic vesicles. NCLs are categorized into different types depending upon the underlying cause i.e., soluble lysosomal enzyme deficiencies or non-enzymatic deficiencies (functions of identified proteins), which are sub-divided based on an axial classification system. In this review, we have evaluated the current evidence in the literature and reported the incidence rates, underlying mechanisms and currently available management protocols for these rare set of neuroophthalmological disorders. Additionally, we also highlighted the potential therapies under development that can expand the treatment of these rare disorders beyond symptomatic relief.
Collapse
Affiliation(s)
- Rohan Bir Singh
- Massachusetts Eye and Ear, Department of Ophthalmology, Harvard Medical School, Boston, MA, USA.,Department of Ophthalmology, Leiden University Medical Center, Leiden, The Netherlands
| | - Prakash Gupta
- Department of Internal Medicine, All India Institute of Medical Sciences, New Delhi, India
| | - Akash Kartik
- Department of Hepatobiliary and Pancreatic Surgery, Mayo Clinic, Rochester, MN, USA
| | - Naba Farooqui
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Sachi Singhal
- Department of Internal Medicine, Mayo Clinic, Rochester, MN, USA
| | - Sukhman Shergill
- Department of Anesthesiology, Yale-New Haven Hospital, New Haven, CT, USA
| | - Kanwar Partap Singh
- Department of Ophthalmology, Dayanand Medical College & Hospital, Ludhiana, India
| | - Aniruddha Agarwal
- Eye Institute, Cleveland Clinic Abu Dhabi, Abu Dhabi, United Arab Emirates
| |
Collapse
|
7
|
Zaghmi A, Drouin-Ouellet J, Brambilla D, Gauthier MA. Treating brain diseases using systemic parenterally-administered protein therapeutics: Dysfunction of the brain barriers and potential strategies. Biomaterials 2020; 269:120461. [PMID: 33218788 DOI: 10.1016/j.biomaterials.2020.120461] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Revised: 09/23/2020] [Accepted: 10/18/2020] [Indexed: 12/12/2022]
Abstract
The parenteral administration of protein therapeutics is increasingly gaining importance for the treatment of human diseases. However, the presence of practically impermeable blood-brain barriers greatly restricts access of such pharmaceutics to the brain. Treating brain disorders with proteins thus remains a great challenge, and the slow clinical translation of these therapeutics may be largely ascribed to the lack of appropriate brain delivery system. Exploring new approaches to deliver proteins to the brain by circumventing physiological barriers is thus of great interest. Moreover, parallel advances in the molecular neurosciences are important for better characterizing blood-brain interfaces, particularly under different pathological conditions (e.g., stroke, multiple sclerosis, Parkinson's disease, and Alzheimer's disease). This review presents the current state of knowledge of the structure and the function of the main physiological barriers of the brain, the mechanisms of transport across these interfaces, as well as alterations to these concomitant with brain disorders. Further, the different strategies to promote protein delivery into the brain are presented, including the use of molecular Trojan horses, the formulation of nanosystems conjugated/loaded with proteins, protein-engineering technologies, the conjugation of proteins to polymers, and the modulation of intercellular junctions. Additionally, therapeutic approaches for brain diseases that do not involve targeting to the brain are presented (i.e., sink and scavenging mechanisms).
Collapse
Affiliation(s)
- A Zaghmi
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC, J3X 1S2, Canada
| | - J Drouin-Ouellet
- Faculty of Pharmacy, Université de Montréal, CP 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - D Brambilla
- Faculty of Pharmacy, Université de Montréal, CP 6128, succ. Centre-ville, Montréal, QC, H3C 3J7, Canada
| | - M A Gauthier
- Institut National de la Recherche Scientifique (INRS), EMT Research Center, Varennes, QC, J3X 1S2, Canada.
| |
Collapse
|
8
|
Coutinho MF, Santos JI, S. Mendonça L, Matos L, Prata MJ, S. Jurado A, Pedroso de Lima MC, Alves S. Lysosomal Storage Disease-Associated Neuropathy: Targeting Stable Nucleic Acid Lipid Particle (SNALP)-Formulated siRNAs to the Brain as a Therapeutic Approach. Int J Mol Sci 2020; 21:ijms21165732. [PMID: 32785133 PMCID: PMC7461213 DOI: 10.3390/ijms21165732] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 07/31/2020] [Accepted: 08/06/2020] [Indexed: 12/22/2022] Open
Abstract
More than two thirds of Lysosomal Storage Diseases (LSDs) present central nervous system involvement. Nevertheless, only one of the currently approved therapies has an impact on neuropathology. Therefore, alternative approaches are under development, either addressing the underlying enzymatic defect or its downstream consequences. Also under study is the possibility to block substrate accumulation upstream, by promoting a decrease of its synthesis. This concept is known as substrate reduction therapy and may be triggered by several molecules, such as small interfering RNAs (siRNAs). siRNAs promote RNA interference, a naturally occurring sequence-specific post-transcriptional gene-silencing mechanism, and may target virtually any gene of interest, inhibiting its expression. Still, naked siRNAs have limited cellular uptake, low biological stability, and unfavorable pharmacokinetics. Thus, their translation into clinics requires proper delivery methods. One promising platform is a special class of liposomes called stable nucleic acid lipid particles (SNALPs), which are characterized by high cargo encapsulation efficiency and may be engineered to promote targeted delivery to specific receptors. Here, we review the concept of SNALPs, presenting a series of examples on their efficacy as siRNA nanodelivery systems. By doing so, we hope to unveil the therapeutic potential of these nanosystems for targeted brain delivery of siRNAs in LSDs.
Collapse
Affiliation(s)
- Maria Francisca Coutinho
- Research and Development Unit, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA I.P), Rua Alexandre Herculano, 321, 4000-055 Porto, Portugal; (J.I.S.); (L.M.); (S.A.)
- Center for the Study of Animal Science, CECA-ICETA, University of Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal
- Correspondence: ; Tel.: +351-(223)-401-113
| | - Juliana Inês Santos
- Research and Development Unit, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA I.P), Rua Alexandre Herculano, 321, 4000-055 Porto, Portugal; (J.I.S.); (L.M.); (S.A.)
- Biology Department, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 4169-007 Porto, Portugal;
| | - Liliana S. Mendonça
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (L.S.M.); (M.C.P.d.L.)
- CIBB—Center for Innovative Biomedicine and Biotechnology, University of Coimbra, 3004-504 Coimbra, Portugal
| | - Liliana Matos
- Research and Development Unit, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA I.P), Rua Alexandre Herculano, 321, 4000-055 Porto, Portugal; (J.I.S.); (L.M.); (S.A.)
- Center for the Study of Animal Science, CECA-ICETA, University of Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal
| | - Maria João Prata
- Biology Department, Faculty of Sciences, University of Porto, Rua do Campo Alegre, 4169-007 Porto, Portugal;
- i3S—Institute of Research and Innovation in Health/IPATIMUP—Institute of Molecular Pathology and Immunology of the University of Porto, Rua Alfredo Allen, 208 4200-135 Porto, Portugal
| | - Amália S. Jurado
- University of Coimbra, CNC—Center for Neuroscience and Cell Biology, Department of Life Sciences, Calçada Martim de Freitas, 3000-456 Coimbra, Portugal;
| | - Maria C. Pedroso de Lima
- CNC—Center for Neuroscience and Cell Biology, University of Coimbra, 3004-504 Coimbra, Portugal; (L.S.M.); (M.C.P.d.L.)
| | - Sandra Alves
- Research and Development Unit, Department of Human Genetics, National Institute of Health Doutor Ricardo Jorge (INSA I.P), Rua Alexandre Herculano, 321, 4000-055 Porto, Portugal; (J.I.S.); (L.M.); (S.A.)
- Center for the Study of Animal Science, CECA-ICETA, University of Porto, Praça Gomes Teixeira, Apartado 55142, 4051-401 Porto, Portugal
| |
Collapse
|
9
|
Favret JM, Weinstock NI, Feltri ML, Shin D. Pre-clinical Mouse Models of Neurodegenerative Lysosomal Storage Diseases. Front Mol Biosci 2020; 7:57. [PMID: 32351971 PMCID: PMC7174556 DOI: 10.3389/fmolb.2020.00057] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2020] [Accepted: 03/20/2020] [Indexed: 12/12/2022] Open
Abstract
There are over 50 lysosomal hydrolase deficiencies, many of which cause neurodegeneration, cognitive decline and death. In recent years, a number of broad innovative therapies have been proposed and investigated for lysosomal storage diseases (LSDs), such as enzyme replacement, substrate reduction, pharmacologic chaperones, stem cell transplantation, and various forms of gene therapy. Murine models that accurately reflect the phenotypes observed in human LSDs are critical for the development, assessment and implementation of novel translational therapies. The goal of this review is to summarize the neurodegenerative murine LSD models available that recapitulate human disease, and the pre-clinical studies previously conducted. We also describe some limitations and difficulties in working with mouse models of neurodegenerative LSDs.
Collapse
Affiliation(s)
| | | | | | - Daesung Shin
- Hunter James Kelly Research Institute, Department of Biochemistry and Neurology, Jacobs School of Medicine and Biomedical Sciences, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
10
|
Nelvagal HR, Cooper JD. An update on the progress of preclinical models for guiding therapeutic management of neuronal ceroid lipofuscinosis. Expert Opin Orphan Drugs 2019. [DOI: 10.1080/21678707.2019.1703672] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- Hemanth Ramesh Nelvagal
- Department of Pediatrics, Division of genetics and genomics, Washington University School of Medicine in St. Louis, St Louis, MO, USA
| | - Jonathan D Cooper
- Department of Pediatrics, Division of genetics and genomics, Washington University School of Medicine in St. Louis, St Louis, MO, USA
| |
Collapse
|
11
|
Gustavsson S, Ohlin Sjöström E, Tjernberg A, Janson J, Westermark U, Andersson T, Makower Å, Arnelöf E, Andersson G, Svartengren J, Ekholm C, Svensson Gelius S. Intravenous delivery of a chemically modified sulfamidase efficiently reduces heparan sulfate storage and brain pathology in mucopolysaccharidosis IIIA mice. Mol Genet Metab Rep 2019; 21:100510. [PMID: 31528541 PMCID: PMC6737345 DOI: 10.1016/j.ymgmr.2019.100510] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Revised: 08/23/2019] [Accepted: 08/23/2019] [Indexed: 01/07/2023] Open
Abstract
Mucopolysaccharidosis type IIIA (MPS IIIA) is a lysosomal storage disorder (LSD) characterized by severe central nervous system (CNS) degeneration. The disease is caused by mutations in the SGSH gene coding for the lysosomal enzyme sulfamidase. Sulfamidase deficiency leads to accumulation of heparan sulfate (HS), which triggers aberrant cellular function, inflammation and eventually cell death. There is currently no available treatment against MPS IIIA. In the present study, a chemically modified recombinant human sulfamidase (CM-rhSulfamidase) with disrupted glycans showed reduced glycan receptor mediated endocytosis, indicating a non-receptor mediated uptake in MPS IIIA patient fibroblasts. Intracellular enzymatic activity and stability was not affected by chemical modification. After intravenous (i.v.) administration in mice, CM-rhSulfamidase showed a prolonged exposure in plasma and distributed to the brain, present both in vascular profiles and in brain parenchyma. Repeated weekly i.v. administration resulted in a dose- and time-dependent reduction of HS in CNS compartments in a mouse model of MPS IIIA. The reduction in HS was paralleled by improvements in lysosomal pathology and neuroinflammation. Behavioral deficits in the MPS IIIA mouse model were apparent in the domains of exploratory behavior, neuromuscular function, social- and learning abilities. CM-rhSulfamidase treatment improved activity in the open field test, endurance in the wire hanging test, sociability in the three-chamber test, whereas other test parameters trended towards improvements. The unique properties of CM-rhSulfamidase described here strongly support the normalization of clinical symptoms, and this candidate drug is therefore currently undergoing clinical studies evaluating safety and efficacy in patients with MPS IIIA.
Collapse
Key Words
- ADA, Anti-drug antibody
- AF, Autofluorescence
- BBB, Blood-brain barrier
- CHO, Chinese hamster ovarian
- CM-rhSulfamidase, Chemically modified recombinant human sulfamidase
- CNS, Central nervous system
- CPM, Chlorpheniramine maleate
- ECL, Electrochemiluminescence
- ERT, Recombinant enzyme replacement therapy
- Enzyme replacement therapy
- GFAP, Glial fibrillary acidic protein
- HS, Heparan sulfate
- Heparan sulfate
- LC-MS, Liquid chromatography-mass spectrometry
- LC-MS/MS, Liquid chromatography-tandem mass spectrometry
- LIMPII, Lysosomal integral membrane protein II
- LSD, Lysosomal storage disease
- M6P, Mannose 6-phosphate
- MPS IIIA, Mucopolysaccharidosis type IIIA
- MSD-ECL, Meso scale discovery electrochemiluminescence
- MTX, Methotrexate
- Mucopolysaccharidosis IIIA
- Neuroinflammation
- PBS, Phosphate buffered saline
- PFA, Paraformaldehyde
- PK, Pharmacokinetic
- RT, Room temperature
- SEC, Size exclusion chromatography
- SEM, Standard error of mean
- Sanfilippo
- Sulfamidase
- TFA, Trifluoroacetic acid
- WT, Wild type
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | | | | | | | - Stefan Svensson Gelius
- Research & Translational Science Unit, Swedish Orphan Biovitrum AB (publ), Stockholm, Sweden
| |
Collapse
|
12
|
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: 3.5] [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
|
13
|
The glycosylation design space for recombinant lysosomal replacement enzymes produced in CHO cells. Nat Commun 2019; 10:1785. [PMID: 31040271 PMCID: PMC6491494 DOI: 10.1038/s41467-019-09809-3] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2018] [Accepted: 03/29/2019] [Indexed: 12/18/2022] Open
Abstract
Lysosomal replacement enzymes are essential therapeutic options for rare congenital lysosomal enzyme deficiencies, but enzymes in clinical use are only partially effective due to short circulatory half-life and inefficient biodistribution. Replacement enzymes are primarily taken up by cell surface glycan receptors, and glycan structures influence uptake, biodistribution, and circulation time. It has not been possible to design and systematically study effects of different glycan features. Here we present a comprehensive gene engineering screen in Chinese hamster ovary cells that enables production of lysosomal enzymes with N-glycans custom designed to affect key glycan features guiding cellular uptake and circulation. We demonstrate distinct circulation time and organ distribution of selected glycoforms of α-galactosidase A in a Fabry disease mouse model, and find that an α2-3 sialylated glycoform designed to eliminate uptake by the mannose 6-phosphate and mannose receptors exhibits improved circulation time and targeting to hard-to-reach organs such as heart. The developed design matrix and engineered CHO cell lines enables systematic studies towards improving enzyme replacement therapeutics. Lysosomal replacement enzymes are taken up by cell surface receptors that recognize glycans, the effects of different glycan features are unknown. Here the authors present a gene engineering screen in CHO cells that allows custom N-glycan-decorated enzymes with improved circulation time and organ distribution.
Collapse
|
14
|
Lu L, Michelena TM, Wong A, Zhang CJ, Meng Y. The inhibition of acetylcholinesterase by a brain-targeting polylysine-ApoE peptide: biochemical and structural characterizations. ANNUAL INTERNATIONAL CONFERENCE OF THE IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. IEEE ENGINEERING IN MEDICINE AND BIOLOGY SOCIETY. ANNUAL INTERNATIONAL CONFERENCE 2018; 2018:155-158. [PMID: 30440362 DOI: 10.1109/embc.2018.8512202] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The in-trans delivery of protein therapeutics across the blood-brain barrier by K16ApoE peptide carrier has been demonstrated to improve the neurological symptoms and increase the life-span of late-infantile neuronal ceroid lipofuscinosis (LINCL) mice. However, acute toxicity of K16ApoE was observed in LINCL mice resulting in a narrow therapeutic index, limiting the potential of translating the K16ApoE into a viable drug delivery system. This study aims to unravel the toxic mechanism of action. We hypothesized that the toxic response towards the peptide was induced by inhibition of acetylcholinesterase (AChE) activity at neuro-muscular junction. Here, results from the dose-response study suggested that AChE activity was inhibited by K16ApoE at either low or high doses but not at the mid-dose where a significant increase in AChE activity was observed. Meanwhile, molecular docking simulations showed that the N-terminus of K16ApoE is capable of binding to the active site gorge of AChE. In addition to a favorable spatial orientation, this docking pose also revealed strong surface charge interactions which may account for the observed inhibitory effect. While statistical analysis of the dose response and survival ratio suggested that AChE is not the primary mechanism of action for the acute toxicity of K16ApoE, both biochemical evidence and structural analysis have assigned indirect but critical roles for AChE in the overall toxicity mechanism of this peptide carrier.
Collapse
|
15
|
Michelena TM, Tian X, Zhou X, Meng Y. The impact on the activity of acetylcholinesterase of a polylysine-ApoE peptide carrier targeting the blood brain barrier. ACTA ACUST UNITED AC 2018. [DOI: 10.2131/fts.5.123] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Toby M. Michelena
- School of Natural, Applied and Health Science, Wenzhou-Kean University, China
| | - Xuechen Tian
- School of Natural, Applied and Health Science, Wenzhou-Kean University, China
| | - Xuan Zhou
- School of Natural, Applied and Health Science, Wenzhou-Kean University, China
| | - Yu Meng
- School of Natural, Applied and Health Science, Wenzhou-Kean University, China
| |
Collapse
|
16
|
Emerging therapies for neuropathic lysosomal storage disorders. Prog Neurobiol 2017; 152:166-180. [DOI: 10.1016/j.pneurobio.2016.10.002] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2015] [Revised: 09/29/2016] [Accepted: 10/02/2016] [Indexed: 12/18/2022]
|
17
|
Meng Y, Wiseman JA, Nemtsova Y, Moore DF, Guevarra J, Reuhl K, Banks WA, Daneman R, Sleat DE, Lobel P. A Basic ApoE-Based Peptide Mediator to Deliver Proteins across the Blood-Brain Barrier: Long-Term Efficacy, Toxicity, and Mechanism. Mol Ther 2017; 25:1531-1543. [PMID: 28456380 DOI: 10.1016/j.ymthe.2017.03.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2016] [Revised: 03/28/2017] [Accepted: 03/29/2017] [Indexed: 11/26/2022] Open
Abstract
We have investigated delivery of protein therapeutics from the bloodstream into the brain using a mouse model of late-infantile neuronal ceroid lipofuscinosis (LINCL), a lysosomal disease due to deficiencies in tripeptidyl peptidase 1 (TPP1). Supraphysiological levels of TPP1 are delivered to the mouse brain by acute intravenous injection when co-administered with K16ApoE, a peptide that in trans mediates passage across the blood-brain barrier (BBB). Chronic treatment of LINCL mice with TPP1 and K16ApoE extended the lifespan from 126 to >294 days, diminished pathology, and slowed locomotor dysfunction. K16ApoE enhanced uptake of a fixable biotin tracer by brain endothelial cells in a dose-dependent manner, suggesting that its mechanism involves stimulation of endocytosis. Pharmacokinetic experiments indicated that K16ApoE functions without disrupting the BBB, with minimal effects on overall clearance or uptake by the liver and kidney. K16ApoE has a narrow therapeutic index, with toxicity manifested as lethargy and/or death in mice. To address this, we evaluated variant peptides but found that efficacy and toxicity are associated, suggesting that desired and adverse effects are mechanistically related. Toxicity currently precludes direct clinical application of peptide-mediated delivery in its present form but it remains a useful approach to proof-of-principle studies for biologic therapies to the brain in animal models.
Collapse
Affiliation(s)
- Yu Meng
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Wenzhou-Kean University, Wenzhou, Zhejiang 32050, China
| | - Jennifer A Wiseman
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Yuliya Nemtsova
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Dirk F Moore
- Department of Biostatistics, School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Jenieve Guevarra
- Department of Pharmacology, Physiology, and Neuroscience, Rutgers New Jersey Medical School, Newark, NJ 07103, USA
| | - Kenneth Reuhl
- Department of Pharmacology and Toxicology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - William A Banks
- Geriatrics Research Education and Clinical Center, Department of Medicine, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; Division of Gerontology and Geriatric Medicine, University of Washington School of Medicine, Seattle, WA 98108, USA
| | - Richard Daneman
- Departments of Pharmacology and Neuroscience, University of California, San Diego, CA 92093, USA
| | - David E Sleat
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Department of Biochemistry and Molecular Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.
| | - Peter Lobel
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Department of Biochemistry and Molecular Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA.
| |
Collapse
|
18
|
Wiseman JA, Meng Y, Nemtsova Y, Matteson PG, Millonig JH, Moore DF, Sleat DE, Lobel P. Chronic Enzyme Replacement to the Brain of a Late Infantile Neuronal Ceroid Lipofuscinosis Mouse Has Differential Effects on Phenotypes of Disease. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2017; 4:204-212. [PMID: 28345005 PMCID: PMC5363315 DOI: 10.1016/j.omtm.2017.01.004] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 01/23/2017] [Indexed: 12/12/2022]
Abstract
Late infantile neuronal ceroid lipofuscinosis (LINCL) is a fatal inherited neurodegenerative disease caused by loss of lysosomal protease tripeptidyl peptidase 1 (TPP1). We have investigated the effects of chronic intrathecal (IT) administration using enzyme replacement therapy (ERT) to the brain of an LINCL mouse model, in which locomotor function declines dramatically prior to early death. Median lifespan was significantly extended from 126 days to >259 days when chronic IT treatment was initiated before the onset of disease. While treated animals lived longer and showed little sign of locomotor dysfunction as measured by stride length, some or all (depending on regimen) still died prematurely. One explanation is that cerebrospinal fluid (CSF)-mediated delivery may not deliver TPP1 to all brain regions. Morphological studies support this, showing delivery of TPP1 to ventral, but not deeper and dorsal regions. When IT treatment is initiated in severely affected LINCL mice, lifespan was extended modestly in most but dramatically extended in approximately one-third of the cohort. Treatment improved locomotor function in these severely compromised animals after it had declined to the point at which animals normally die. This indicates that some pathology in LINCL is reversible and does not simply reflect neuronal death.
Collapse
Affiliation(s)
- Jennifer A Wiseman
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Yu Meng
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Yuliya Nemtsova
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Paul G Matteson
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - James H Millonig
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Department of Neuroscience & Cell Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Dirk F Moore
- School of Public Health, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - David E Sleat
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Department of Biochemistry and Molecular Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| | - Peter Lobel
- Center for Advanced Biotechnology and Medicine, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA; Department of Biochemistry and Molecular Biology, Rutgers, The State University of New Jersey, Piscataway, NJ 08854, USA
| |
Collapse
|
19
|
Geraets RD, Koh SY, Hastings ML, Kielian T, Pearce DA, Weimer JM. Moving towards effective therapeutic strategies for Neuronal Ceroid Lipofuscinosis. Orphanet J Rare Dis 2016; 11:40. [PMID: 27083890 PMCID: PMC4833901 DOI: 10.1186/s13023-016-0414-2] [Citation(s) in RCA: 57] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2015] [Accepted: 03/16/2016] [Indexed: 12/24/2022] Open
Abstract
The Neuronal Ceroid Lipofuscinoses (NCLs) are a family of autosomal recessive neurodegenerative disorders that annually affect 1:100,000 live births worldwide. This family of diseases results from mutations in one of 14 different genes that share common clinical and pathological etiologies. Clinically, the diseases are subcategorized into infantile, late-infantile, juvenile and adult forms based on their age of onset. Though the disease phenotypes may vary in their age and order of presentation, all typically include progressive visual deterioration and blindness, cognitive impairment, motor deficits and seizures. Pathological hallmarks of NCLs include the accumulation of storage material or ceroid in the lysosome, progressive neuronal degeneration and massive glial activation. Advances have been made in genetic diagnosis and counseling for families. However, comprehensive treatment programs that delay or halt disease progression have been elusive. Current disease management is primarily targeted at controlling the symptoms rather than "curing" the disease. Recognizing the growing need for transparency and synergistic efforts to move the field forward, this review will provide an overview of the therapeutic approaches currently being pursued in preclinical and clinical trials to treat different forms of NCL as well as provide insight to novel therapeutic approaches in development for the NCLs.
Collapse
Affiliation(s)
- Ryan D. Geraets
- />Children’s Health Research Center, Sanford Research, Sioux Falls, SD USA
- />Sanford School of Medicine at the University of South Dakota, Sioux Falls, SD USA
| | - Seung yon Koh
- />Children’s Health Research Center, Sanford Research, Sioux Falls, SD USA
| | - Michelle L. Hastings
- />Department of Cell Biology and Anatomy, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, IL USA
| | - Tammy Kielian
- />Department of Pathology and Microbiology, University of Nebraska Medical Center, Omaha, NE USA
| | - David A. Pearce
- />Children’s Health Research Center, Sanford Research, Sioux Falls, SD USA
- />Sanford School of Medicine at the University of South Dakota, Sioux Falls, SD USA
| | - Jill M. Weimer
- />Children’s Health Research Center, Sanford Research, Sioux Falls, SD USA
- />Sanford School of Medicine at the University of South Dakota, Sioux Falls, SD USA
| |
Collapse
|
20
|
Damme M, Stroobants S, Lüdemann M, Rothaug M, Lüllmann-Rauch R, Beck HC, Ericsson A, Andersson C, Fogh J, D'Hooge R, Saftig P, Blanz J. Chronic enzyme replacement therapy ameliorates neuropathology in alpha-mannosidosis mice. Ann Clin Transl Neurol 2015; 2:987-1001. [PMID: 26817023 PMCID: PMC4693626 DOI: 10.1002/acn3.245] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2015] [Revised: 08/03/2015] [Accepted: 08/03/2015] [Indexed: 01/17/2023] Open
Abstract
OBJECTIVE The lysosomal storage disease alpha-mannosidosis is caused by the deficiency of the lysosomal acid hydrolase alpha-mannosidase (LAMAN) leading to lysosomal accumulation of neutral mannose-linked oligosaccharides throughout the body, including the brain. Clinical findings in alpha-mannosidosis include skeletal malformations, intellectual disabilities and hearing impairment. To date, no curative treatment is available. We previously developed a beneficial enzyme replacement therapy (ERT) regimen for alpha-mannosidase knockout mice, a valid mouse model for the human disease. However, humoral immune responses against the injected recombinant human alpha-mannosidase (rhLAMAN) precluded long-term studies and chronic treatment. METHODS Here, we describe the generation of an immune-tolerant alpha-mannosidosis mouse model that allowed chronic injection of rhLAMAN by transgenic expression of a catalytically inactive variant of human LAMAN in the knockout background. RESULTS Chronic ERT of rhLAMAN revealed pronounced effects on primary substrate storage throughout the brain, normalization of lysosomal enzyme activities and morphology as well as a decrease in microglia activation. The positive effect of long-term ERT on neuronal lysosomal function was reflected by an improvement of cognitive deficits and exploratory activity. in vivo and in vitro uptake measurements indicate rapid clearance of rhLAMAN from circulation and a broad uptake into different cell types of the nervous system. INTERPRETATION Our data contribute to the understanding of neurological disorders treatment by demonstrating that lysosomal enzymes such as rhLAMAN can penetrate into the brain and is able to ameliorate neuropathology.
Collapse
Affiliation(s)
- Markus Damme
- Biochemical Institute University of Kiel D-24098 Kiel Germany
| | - Stijn Stroobants
- Laboratory of Biological Psychology University of Leuven B-3000 Leuven Belgium
| | - Meike Lüdemann
- Biochemical Institute University of Kiel D-24098 Kiel Germany
| | | | | | - Hans Christian Beck
- Department of Biochemistry and Pharmacology Centre for Clinical Proteomics Odense University Hospital Sdr Boulevard 29 DK-5000 Odense C Denmark
| | | | | | - Jens Fogh
- Zymenex A/S Roskildevej 12C 3400 Hillerød Denmark
| | - Rudi D'Hooge
- Laboratory of Biological Psychology University of Leuven B-3000 Leuven Belgium
| | - Paul Saftig
- Biochemical Institute University of Kiel D-24098 Kiel Germany
| | - Judith Blanz
- Biochemical Institute University of Kiel D-24098 Kiel Germany
| |
Collapse
|
21
|
|
22
|
Cooper JD, Tarczyluk MA, Nelvagal HR. Towards a new understanding of NCL pathogenesis. Biochim Biophys Acta Mol Basis Dis 2015; 1852:2256-61. [PMID: 26026924 DOI: 10.1016/j.bbadis.2015.05.014] [Citation(s) in RCA: 57] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2015] [Revised: 05/20/2015] [Accepted: 05/21/2015] [Indexed: 01/29/2023]
Abstract
The Neuronal Ceroid Lipofuscinoses (NCLs, Batten disease) are a group of inherited neurodegenerative disorders that have been traditionally grouped together on the basis of certain shared clinical and pathological features. However, as the number of genes that appear to cause new forms of NCL continues to grow, it is timely to reassess our understanding of the pathogenesis of these disorders and what groups them together. The various NCL subtypes do indeed share features of a build-up of autofluorescent storage material, progressive neuron loss and activation of the innate immune system. The characterisation of animal models has highlighted the selective nature of neuron loss and its intimate relationship with glial activation, rather than the generalised build-up of storage material. More recent data provide evidence for the pathway-dependent nature of pathology, the contribution of glial dysfunction, and the involvement of new brain regions previously thought to be unaffected, and it is becoming apparent that pathology extends beyond the brain. These data have important implications, not just for therapy, but also for our understanding of these disorders. However, looking beneath these broadly similar pathological themes evidence emerges for marked differences in the nature and extent of these events in different forms of NCL. Indeed, given the widely different nature of the mutated gene products it is perhaps more surprising that these disorders resemble each other as much as they do. Such data raise the question whether we should rethink the collective grouping of these gene deficiencies together, or whether it would be better to consider them as separate entities. This article is part of a Special Issue entitled: Current Research on the Neuronal Ceroid Lipofuscinoses (Batten Disease).
Collapse
Affiliation(s)
- Jonathan D Cooper
- Pediatric Storage Disorders Laboratory (PSDL), Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology & Neuroscience, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK.
| | - Marta A Tarczyluk
- Pediatric Storage Disorders Laboratory (PSDL), Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology & Neuroscience, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| | - Hemanth R Nelvagal
- Pediatric Storage Disorders Laboratory (PSDL), Department of Basic and Clinical Neuroscience, King's College London, Institute of Psychiatry, Psychology & Neuroscience, James Black Centre, 125 Coldharbour Lane, London SE5 9NU, UK
| |
Collapse
|
23
|
Hassiotis S, Beard H, Luck A, Trim PJ, King B, Snel MF, Hopwood JJ, Hemsley KM. Disease stage determines the efficacy of treatment of a paediatric neurodegenerative disease. Eur J Neurosci 2015; 39:2139-50. [PMID: 25068161 DOI: 10.1111/ejn.12557] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
Lysosomal storage disorders are a large group of inherited metabolic conditions resulting from the deficiency of proteins involved in lysosomal catabolism, with resulting accumulation of substrates inside the cell. Two-thirds of these disorders are associated with a neurodegenerative phenotype and, although few therapeutic options are available to patients at present, clinical trials of several treatments including lysosomal enzyme replacement are underway. Although animal studies indicate the efficacy of presymptomatic treatment, it is largely unknown whether symptomatic disease-related pathology and functional deficits are reversible. To begin to address this, we used a naturally-occurring mouse model with Sanfilippo syndrome (mucopolysaccharidosis type IIIA) to examine the effectiveness of intracisternal cerebrospinal fluid enzyme replacement in early, mid- and symptomatic disease stage mice. We observed a disease-stage-dependent treatment effect, with the most significant reductions in primary and secondary substrate accumulation, astrogliosis and protein aggregate accumulation seen in mucopolysaccharidosis type IIIA mice treated very early in the disease course. Affected mice treated at a symptomatic age exhibited little change in these neuropathological markers in the time-frame of the study. Microgliosis was refractory to treatment regardless of the age at which treatment was instigated. Although longer-term studies are warranted, these findings indicate the importance of early intervention in this condition.
Collapse
|
24
|
A Hitchhiker's guide to the blood-brain barrier: in trans delivery of a therapeutic enzyme. Mol Ther 2014; 22:483-484. [PMID: 24584077 DOI: 10.1038/mt.2014.12] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
|
25
|
Effective intravenous therapy for neurodegenerative disease with a therapeutic enzyme and a peptide that mediates delivery to the brain. Mol Ther 2013; 22:547-553. [PMID: 24394185 DOI: 10.1038/mt.2013.267] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2013] [Accepted: 11/20/2013] [Indexed: 12/20/2022] Open
Abstract
The blood-brain barrier (BBB) presents a major challenge to effective treatment of neurological disorders, including lysosomal storage diseases (LSDs), which frequently present with life-shortening and untreatable neurodegeneration. There is considerable interest in methods for intravenous delivery of lysosomal proteins across the BBB but for the most part, levels achievable in the brain of mouse models are modest and increased lifespan remains to be demonstrated. In this study, we have investigated delivery across the BBB using a mouse model of late-infantile neuronal ceroid lipofuscinosis (LINCL), a neurodegenerative LSD caused by loss of tripeptidyl peptidase I (TPP1). We have achieved supraphysiological levels of TPP1 throughout the brain of LINCL mice by intravenous (IV) coadministration of recombinant TPP1 with a 36-residue peptide that contains polylysine and a low-density lipoprotein receptor binding sequence from apolipoprotein E. Importantly, IV administration of TPP1 with the peptide significantly reduces brain lysosomal storage, increases lifespan and improves neurological function. This simple "mix and inject" method is immediately applicable towards evaluation of enzyme replacement therapy to the brain in preclinical models and further exploration of its clinical potential is warranted.
Collapse
|
26
|
Sondhi D, Rosenberg JB, Van de Graaf BG, Kaminsky SM, Crystal RG. Advances in the treatment of neuronal ceroid lipofuscinosis. Expert Opin Orphan Drugs 2013. [DOI: 10.1517/21678707.2013.852081] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
|
27
|
Sly WS, Vogler C. The final frontier -- crossing the blood-brain barrier. EMBO Mol Med 2013; 5:655-7. [PMID: 23653302 PMCID: PMC3662309 DOI: 10.1002/emmm.201302668] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2013] [Revised: 03/14/2013] [Accepted: 03/15/2013] [Indexed: 11/07/2022] Open
Affiliation(s)
- William S Sly
- Edward A. Doisy Department of Biochemistry and Molecular Biology, Saint Louis University School of Medicine, St. Louis, MO, USA.
| | | |
Collapse
|
28
|
Hawkins-Salsbury JA, Cooper JD, Sands MS. Pathogenesis and therapies for infantile neuronal ceroid lipofuscinosis (infantile CLN1 disease). Biochim Biophys Acta Mol Basis Dis 2013; 1832:1906-9. [PMID: 23747979 DOI: 10.1016/j.bbadis.2013.05.026] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2013] [Revised: 05/08/2013] [Accepted: 05/09/2013] [Indexed: 11/17/2022]
Abstract
The neuronal ceroid lipofuscinoses (NCL, Batten disease) are a group of inherited neurodegenerative diseases. Infantile neuronal ceroid lipofuscinosis (INCL, infantile Batten disease, or infantile CLN1 disease) is caused by a deficiency in the soluble lysosomal enzyme palmitoyl protein thioesterase-1 (PPT1) and has the earliest onset and fastest progression of all the NCLs. Several therapeutic strategies including enzyme replacement, gene therapy, stem cell-mediated therapy, and small molecule drugs have resulted in minimal to modest improvements in the murine model of PPT1-deficiency. However, more recent studies using various combinations of these approaches have shown more promising results; in some instances more than doubling the lifespan of PPT1-deficient mice. These combination therapies that target different pathogenic mechanisms may offer the hope of treating this profoundly neurodegenerative disorder. Similar approaches may be useful when treating other forms of NCL caused by deficiencies in soluble lysosomal proteins. Different therapeutic targets will need to be identified and novel strategies developed in order to effectively treat forms of NCL caused by deficiencies in integral membrane proteins such as juvenile neuronal ceroid lipofuscinosis. Finally, the challenge with all of the NCLs will lie in early diagnosis, improving the efficacy of the treatments, and effectively translating them into the clinic. This article is part of a Special Issue entitled: The Neuronal Ceroid Lipofuscinoses or Batten Disease.
Collapse
Affiliation(s)
- Jacqueline A Hawkins-Salsbury
- Washington University School of Medicine, Department of Internal Medicine, Box 8007, 660 South Euclid Avenue, St. Louis, MO 63110, USA
| | | | | |
Collapse
|
29
|
Biochemical evidence for superior correction of neuronal storage by chemically modified enzyme in murine mucopolysaccharidosis VII. Proc Natl Acad Sci U S A 2012; 109:17022-7. [PMID: 23027951 DOI: 10.1073/pnas.1214779109] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Enzyme replacement therapy has been used successfully in many lysosomal storage diseases. However, correction of brain storage has been limited by the inability of infused enzyme to cross the blood-brain barrier (BBB). We recently reported that PerT-GUS, a form of β-glucuronidase (GUS) chemically modified to eliminate its uptake and clearance by carbohydrate-dependent receptors, crossed the BBB and cleared neuronal storage in an immunotolerant model of murine mucopolysaccharidosis (MPS) type VII. In this respect, the chemically modified enzyme was superior to native β-glucuronidase. Chemically modified enzyme was also delivered more effectively to heart, kidney, and muscle. However, liver and spleen, which express high levels of carbohydrate receptors, received nearly fourfold lower levels of PerT-GUS compared with native GUS. A recent report on PerT-treated sulfamidase in murine MPS IIIA confirmed enhanced delivery to other tissues but failed to observe clearance of storage in neurons. To confirm and extend our original observations, we compared the efficacy of 12 weekly i.v. infusions of PerT-GUS versus native GUS on (i) delivery of enzyme to brain; (ii) improvement in histopathology; and (iii) correction of secondary elevations of other lysosomal enzymes. Such correction is a recognized biomarker for correction of neuronal storage. PerT-GUS was superior to native GUS in all three categories. These results provide additional evidence that long-circulating enzyme, chemically modified to escape carbohydrate-mediated clearance, may offer advantages in treating MPS VII. The relevance of this approach to treat other lysosomal storage diseases that affect brain awaits confirmation.
Collapse
|