1
|
Antipova V, Heimes D, Seidel K, Schulz J, Schmitt O, Holzmann C, Rolfs A, Bidmon HJ, González de San Román Martín E, Huesgen PF, Amunts K, Keiler J, Hammer N, Witt M, Wree A. Differently increased volumes of multiple brain areas in Npc1 mutant mice following various drug treatments. Front Neuroanat 2024; 18:1430790. [PMID: 39081805 PMCID: PMC11286580 DOI: 10.3389/fnana.2024.1430790] [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: 05/10/2024] [Accepted: 07/01/2024] [Indexed: 08/02/2024] Open
Abstract
Background Niemann-Pick disease type C1 (NPC1, MIM 257220) is a heritable lysosomal storage disease characterized by a progressive neurological degeneration that causes disability and premature death. A murine model of Npc1-/- displays a rapidly progressing form of Npc1 disease, which is characterized by weight loss, ataxia, and increased cholesterol storage. Npc1-/- mice receiving a combined therapy (COMBI) of miglustat (MIGLU), the neurosteroid allopregnanolone (ALLO) and the cyclic oligosaccharide 2-hydroxypropyl-β-cyclodextrin (HPßCD) showed prevention of Purkinje cell loss, improved motor function and reduced intracellular lipid storage. Although therapy of Npc1-/- mice with COMBI, MIGLU or HPßCD resulted in the prevention of body weight loss, reduced total brain weight was not positively influenced. Methods In order to evaluate alterations of different brain areas caused by pharmacotherapy, fresh volumes (volumes calculated from the volumes determined from paraffin embedded brain slices) of various brain structures in sham- and drug-treated wild type and mutant mice were measured using stereological methods. Results In the wild type mice, the volumes of investigated brain areas were not significantly altered by either therapy. Compared with the respective wild types, fresh volumes of specific brain areas, which were significantly reduced in sham-treated Npc1-/- mice, partly increased after the pharmacotherapies in all treatment strategies; most pronounced differences were found in the CA1 area of the hippocampus and in olfactory structures. Discussion Volumes of brain areas of Npc1-/- mice were not specifically changed in terms of functionality after administering COMBI, MIGLU, or HPßCD. Measurements of fresh volumes of brain areas in Npc1-/- mice could monitor region-specific changes and response to drug treatment that correlated, in part, with behavioral improvements in this mouse model.
Collapse
Affiliation(s)
- Veronica Antipova
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
- Division of Macroscopic and Clinical Anatomy, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz, Austria
| | - Diana Heimes
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
- Department of Oral and Maxillofacial Surgery, University Medical Center Mainz, Mainz, Germany
| | - Katharina Seidel
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
- Klinik für Frauenheilkunde und Geburtshilfe, Dietrich-Bonhoeffer-Klinikum, Neubrandenburg, Germany
| | - Jennifer Schulz
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Oliver Schmitt
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
- Department of Anatomy, Medical School Hamburg, University of Applied Sciences and Medical University, Hamburg, Germany
| | - Carsten Holzmann
- Institute of Medical Genetics, Rostock University Medical Center, Rostock, Germany
- Centre of Transdisciplinary Neuroscience Rostock, Rostock, Germany
| | - Arndt Rolfs
- Medical Faculty, University of Rostock, Rostock, Germany
| | - Hans-Jürgen Bidmon
- Institute of Neurosciences and Medicine, Structural and Functional Organisation of the Brain (INM-1), Forschungszentrum Jülich, Jülich, Germany
- Central Institute of Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, Jülich, Germany
| | | | - Pitter F. Huesgen
- Central Institute of Engineering, Electronics and Analytics, ZEA-3, Forschungszentrum Jülich, Jülich, Germany
- Institut für Biologie II, AG Funktional Proteomics, Freiburg, Germany
| | - Katrin Amunts
- Institute of Neurosciences and Medicine, Structural and Functional Organisation of the Brain (INM-1), Forschungszentrum Jülich, Jülich, Germany
- C. and O. Vogt Institute for Brain Research, University Hospital Düsseldorf, University Düsseldorf, Düsseldorf, Germany
| | - Jonas Keiler
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
| | - Niels Hammer
- Division of Macroscopic and Clinical Anatomy, Gottfried Schatz Research Center for Cell Signaling, Metabolism and Aging, Medical University of Graz, Graz, Austria
- Department of Orthopedic and Trauma Surgery, University of Leipzig, Leipzig, Germany
- Division of Biomechatronics, Fraunhofer Institute for Machine Tools and Forming Technology, Dresden, Germany
| | - Martin Witt
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
- Department of Anatomy, Technische Universität Dresden, Dresden, Germany
- Department of Anatomy, Institute of Biostructural Basics of Medical Sciences, Poznan Medical University, Poznan, Poland
| | - Andreas Wree
- Institute of Anatomy, Rostock University Medical Center, Rostock, Germany
- Centre of Transdisciplinary Neuroscience Rostock, Rostock, Germany
| |
Collapse
|
2
|
Cologna SM, Pathmasiri KC, Pergande MR, Rosenhouse-Dantsker A. Alterations in Cholesterol and Phosphoinositides Levels in the Intracellular Cholesterol Trafficking Disorder NPC. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2023; 1422:143-165. [PMID: 36988880 DOI: 10.1007/978-3-031-21547-6_5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 03/30/2023]
Abstract
Lipid mistrafficking is a biochemical hallmark of Niemann-Pick Type C (NPC) disease and is classically characterized with endo/lysosomal accumulation of unesterified cholesterol due to genetic mutations in the cholesterol transporter proteins NPC1 and NPC2. Storage of this essential signaling lipid leads to a sequence of downstream events, including oxidative stress, calcium imbalance, neuroinflammation, and progressive neurodegeneration, another hallmark of NPC disease. These observations have been validated in a growing number of studies ranging from NPC cell cultures and animal models to patient specimens. In recent reports, alterations in the levels of another class of critical signaling lipids, namely phosphoinositides, have been described in NPC disease. Focusing on cholesterol and phosphoinositides, the chapter begins by reviewing the interactions of NPC proteins with cholesterol and their role in cholesterol transport. It then continues to describe the modulation of cholesterol efflux in NPC disease. The chapter concludes with a summary of findings related to the functional consequences of perturbations in phosphoinositides in this fatal disease.
Collapse
Affiliation(s)
| | | | - Melissa R Pergande
- Department of Chemistry, University of Illinois Chicago, Chicago, IL, USA
| | | |
Collapse
|
3
|
Organ Weights in NPC1 Mutant Mice Partly Normalized by Various Pharmacological Treatment Approaches. Int J Mol Sci 2022; 24:ijms24010573. [PMID: 36614015 PMCID: PMC9820376 DOI: 10.3390/ijms24010573] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/21/2022] [Revised: 12/14/2022] [Accepted: 12/22/2022] [Indexed: 12/31/2022] Open
Abstract
Niemann-Pick Type C1 (NPC1, MIM 257220) is a rare, progressive, lethal, inherited autosomal-recessive endolysosomal storage disease caused by mutations in the NPC1 leading to intracellular lipid storage. We analyzed mostly not jet known alterations of the weights of 14 different organs in the BALB/cNctr-Npc1m1N/-J Jackson Npc1 mice in female and male Npc1+/+ and Npc1-/- mice under various treatment strategies. Mice were treated with (i) no therapy, (ii) vehicle injection, (iii) a combination of miglustat, allopregnanolone, and 2-hydroxypropyl-ß-cyclodextrin (HPßCD), (iv) miglustat, and (v) HPßCD alone starting at P7 and repeated weekly throughout life. The 12 respective male and female wild-type mice groups were evaluated in parallel. In total, 351 mice (176 Npc1+/+, 175 Npc1-/-) were dissected at P65. In both sexes, the body weights of None and Sham Npc1-/- mice were lower than those of respective Npc1+/+ mice. The influence of the Npc1 mutation and/or sex on the weights of various organs, however, differed considerably. In males, Npc1+/+ and Npc1-/- mice had comparable absolute weights of lungs, spleen, and adrenal glands. In Npc1-/- mice, smaller weights of hearts, livers, kidneys, testes, vesicular, and scent glands were found. In female Npc1-/- mice, ovaries, and uteri were significantly smaller. In Npc1-/- mice, relative organ weights, i.e., normalized with body weights, were sex-specifically altered to different extents by the different therapies. The combination of miglustat, allopregnanolone, and the sterol chelator HPßCD partly normalized the weights of more organs than miglustat or HPßCD mono-therapies.
Collapse
|
4
|
Ishitsuka Y, Irie T, Matsuo M. Cyclodextrins applied to the treatment of lysosomal storage disorders. Adv Drug Deliv Rev 2022; 191:114617. [PMID: 36356931 DOI: 10.1016/j.addr.2022.114617] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 09/14/2022] [Accepted: 11/02/2022] [Indexed: 11/09/2022]
Abstract
Cyclodextrin (CD), a cyclic oligosaccharide, is a pharmaceutical additive that improves the solubility of hydrophobic compounds. Recent research has focused on the potential active pharmaceutical abilities of CD. Lysosomal storage diseases are inherited metabolic diseases characterized by lysosomal dysfunction and abnormal lipid storage. Niemann-Pick disease type C (NPC) is caused by mutations in cholesterol transporter genes (NPC1, NPC2) and is characterized by cholesterol accumulation in lysosomes. A biocompatible cholesterol solubilizer 2-hydroxypropyl-β-cyclodextrin (HP-β-CD) was recently used in NPC patients for compassionate use and in clinical trials. HP-β-CD is an attractive drug candidate for NPC; however, its adverse effects, such as ototoxicity, should be solved. In this review, we discuss the current use of HP-β-CD in basic and clinical research and discuss alternative CD derivatives that may outperform HP-β-CD, which should be considered for clinical use. The potential of CD therapy for the treatment of other lysosomal storage diseases is also discussed.
Collapse
Affiliation(s)
- Yoichi Ishitsuka
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan.
| | - Tetsumi Irie
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan; Department of Pharmaceutical Packaging Technology, Faculty of Life Sciences, Kumamoto University, 5-1 Oe-honmachi, Chuo-ku, Kumamoto 862-0973, Japan
| | - Muneaki Matsuo
- Department of Pediatrics, Faculty of Medicine, Saga University, 5-1-1, Nabeshima, Saga 849-8501, Japan
| |
Collapse
|
5
|
Zhang C, Ding D, Sun W, Hu BH, Manohar S, Salvi R. Time- and frequency-dependent changes in acoustic startle reflex amplitude following cyclodextrin-induced outer and inner cell loss. Hear Res 2022; 415:108441. [DOI: 10.1016/j.heares.2022.108441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/04/2021] [Revised: 01/04/2022] [Accepted: 01/13/2022] [Indexed: 11/27/2022]
|
6
|
Becktel DA, Zbesko JC, Frye JB, Chung AG, Hayes M, Calderon K, Grover JW, Li A, Garcia FG, Tavera-Garcia MA, Schnellmann RG, Wu HJJ, Nguyen TVV, Doyle KP. Repeated Administration of 2-Hydroxypropyl-β-Cyclodextrin (HPβCD) Attenuates the Chronic Inflammatory Response to Experimental Stroke. J Neurosci 2022; 42:325-348. [PMID: 34819339 PMCID: PMC8802936 DOI: 10.1523/jneurosci.0933-21.2021] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/03/2021] [Revised: 08/24/2021] [Accepted: 11/16/2021] [Indexed: 11/30/2022] Open
Abstract
Globally, more than 67 million people are living with the effects of ischemic stroke. Importantly, many stroke survivors develop a chronic inflammatory response that may contribute to cognitive impairment, a common and debilitating sequela of stroke that is insufficiently studied and currently untreatable. 2-Hydroxypropyl-β-cyclodextrin (HPβCD) is an FDA-approved cyclic oligosaccharide that can solubilize and entrap lipophilic substances. The goal of the present study was to determine whether the repeated administration of HPβCD curtails the chronic inflammatory response to stroke by reducing lipid accumulation within stroke infarcts in a distal middle cerebral artery occlusion mouse model of stroke. To achieve this goal, we subcutaneously injected young adult and aged male mice with vehicle or HPβCD 3 times per week, with treatment beginning 1 week after stroke. We evaluated mice at 7 weeks following stroke using immunostaining, RNA sequencing, lipidomic, and behavioral analyses. Chronic stroke infarct and peri-infarct regions of HPβCD-treated mice were characterized by an upregulation of genes involved in lipid metabolism and a downregulation of genes involved in innate and adaptive immunity, reactive astrogliosis, and chemotaxis. Correspondingly, HPβCD reduced the accumulation of lipid droplets, T lymphocytes, B lymphocytes, and plasma cells in stroke infarcts. Repeated administration of HPβCD also preserved NeuN immunoreactivity in the striatum and thalamus and c-Fos immunoreactivity in hippocampal regions. Additionally, HPβCD improved recovery through the protection of hippocampal-dependent spatial working memory and reduction of impulsivity. These results indicate that systemic HPβCD treatment following stroke attenuates chronic inflammation and secondary neurodegeneration and prevents poststroke cognitive decline.SIGNIFICANCE STATEMENT Dementia is a common and debilitating sequela of stroke. Currently, there are no available treatments for poststroke dementia. Our study shows that lipid metabolism is disrupted in chronic stroke infarcts, which causes an accumulation of uncleared lipid debris and correlates with a chronic inflammatory response. To our knowledge, these substantial changes in lipid homeostasis have not been previously recognized or investigated in the context of ischemic stroke. We also provide a proof of principle that solubilizing and entrapping lipophilic substances using HPβCD could be an effective strategy for treating chronic inflammation after stroke and other CNS injuries. We propose that using HPβCD for the prevention of poststroke dementia could improve recovery and increase long-term quality of life in stroke sufferers.
Collapse
Affiliation(s)
- Danielle A Becktel
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Jacob C Zbesko
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Jennifer B Frye
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Amanda G Chung
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Megan Hayes
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Kylie Calderon
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | - Jeffrey W Grover
- Department of Molecular and Cellular Biology, University of Arizona, Tucson, Arizona 85719
| | - Anna Li
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
- Arizona Arthritis Center, University of Arizona, Tucson, Arizona 85719
| | - Frankie G Garcia
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
| | | | - Rick G Schnellmann
- Department of Pharmacology and Toxicology, University of Arizona, Tucson, Arizona 85719
| | - Hsin-Jung Joyce Wu
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
- Arizona Arthritis Center, University of Arizona, Tucson, Arizona 85719
| | - Thuy-Vi V Nguyen
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
- Department of Neurology, University of Arizona, Tucson, Arizona 85719
| | - Kristian P Doyle
- Department of Immunobiology, University of Arizona, Tucson, Arizona 85719
- Department of Neurology, University of Arizona, Tucson, Arizona 85719
- BIO5 Institute, University of Arizona, Tucson, Arizona 85719
- Arizona Center on Aging, University of Arizona, Tucson, Arizona 85719
- Department of Psychology, University of Arizona, Tucson, Arizona 85719
- Department of Neurosurgery, University of Arizona, Tucson, Arizona 85719
| |
Collapse
|
7
|
Aqul AA, Ramirez CM, Lopez AM, Burns DK, Repa JJ, Turley SD. Molecular markers of brain cholesterol homeostasis are unchanged despite a smaller brain mass in a mouse model of cholesteryl ester storage disease. Lipids 2022; 57:3-16. [PMID: 34618372 PMCID: PMC8766890 DOI: 10.1002/lipd.12325] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2021] [Revised: 09/17/2021] [Accepted: 09/20/2021] [Indexed: 01/03/2023]
Abstract
Lysosomal acid lipase (LAL), encoded by the gene LIPA, facilitates the intracellular processing of lipids by hydrolyzing cholesteryl esters and triacylglycerols present in newly internalized lipoproteins. Loss-of-function mutations in LIPA result in cholesteryl ester storage disease (CESD) or Wolman disease when mutations cause complete loss of LAL activity. Although the phenotype of a mouse CESD model has been extensively characterized, there has not been a focus on the brain at different stages of disease progression. In the current studies, whole-brain mass and the concentrations of cholesterol in both the esterified (EC) and unesterified (UC) fractions were measured in Lal-/- and matching Lal+/+ mice (FVB-N strain) at ages ranging from 14 up to 280 days after birth. Compared to Lal+/+ controls at 50, 68-76, 140-142, and 230-280 days of age, Lal-/- mice had brain weights that averaged approximately 6%, 7%, 18%, and 20% less, respectively. Brain EC levels were higher in the Lal-/- mice at every age, being elevated 27-fold at 230-280 days. Brain UC concentrations did not show a genotypic difference at any age. The elevated brain EC levels in the Lal-/- mice did not reflect EC in residual blood. An mRNA expression analysis for an array of genes involved in the synthesis, catabolism, storage, and transport of cholesterol in the brains of 141-day old mice did not detect any genotypic differences although the relative mRNA levels for several markers of inflammation were moderately elevated in the Lal-/- mice. The possible sites of EC accretion in the central nervous system are discussed.
Collapse
Affiliation(s)
- Amal A. Aqul
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas TX 75390 USA
| | - Charina M. Ramirez
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas TX 75390 USA
| | - Adam M. Lopez
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas TX 75390 USA
| | - Dennis K. Burns
- Department of Pathology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas TX 75390 USA
| | - Joyce J. Repa
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas TX 75390 USA
- Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas TX 75390 USA
| | - Stephen D. Turley
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd., Dallas TX 75390 USA
| |
Collapse
|
8
|
Manohar S, Ding D, Jiang H, Li L, Chen GD, Kador P, Salvi R. Combined antioxidants and anti-inflammatory therapies fail to attenuate the early and late phases of cyclodextrin-induced cochlear damage and hearing loss. Hear Res 2021; 414:108409. [PMID: 34953289 DOI: 10.1016/j.heares.2021.108409] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 09/08/2021] [Accepted: 12/06/2021] [Indexed: 11/04/2022]
Abstract
Niemann-Pick C1 (NPC1) is a fatal neurodegenerative disease caused by aberrant cholesterol metabolism. The progression of the disease can be slowed by removing excess cholesterol with high-doses of 2-hyroxypropyl-beta-cyclodextrin (HPβCD). Unfortunately, HPβCD causes hearing loss; the initial first phase involves a rapid destruction of outer hair cells (OHCs) while the second phase, occurring 4-6 weeks later, involves the destruction of inner hair cells (IHCs), pillar cells, collapse of the organ of Corti and spiral ganglion neuron degeneration. To determine whether the first and/or second phase of HPβCD-induced cochlear damage is linked, in part, to excess oxidative stress or neuroinflammation, rats were treated with a single-dose of 3000 mg/kg HPβCD alone or together with one of two combination therapies. Each combination therapy was administered from 2-days before to 6-weeks after the HPβCD treatment. Combination 1 consisted of minocycline, an antibiotic that suppresses neuroinflammation, and HK-2, a multifunctional redox modulator that suppresses oxidative stress. Combination 2 was comprised of minocycline plus N-acetyl cysteine (NAC), which upregulates glutathione, a potent antioxidant. To determine if either combination therapy could prevent HPβCD-induced hearing impairment and cochlear damage, distortion product otoacoustic emissions (DPOAE) were measured to assess OHC function and the cochlear compound action potential (CAP) was measured to assess the function of IHCs and auditory nerve fibers. Cochleograms were prepared to quantify the amount of OHC, IHC and pillar cell (PC) loss. HPβCD significantly reduced DPOAE and CAP amplitudes and caused significant OHC, IHC and OPC losses with losses greater in the high-frequency base of the cochlea than the apex. Neither minocycline + HK-2 (MIN+ HK-2) nor minocycline + NAC (MIN+NAC) prevented the loss of DPOAEs, CAPs, OHCs, IHCs or IPCs caused by HPβCD. These results suggest that oxidative stress and neuroinflammation are unlikely to play major roles in mediating the first or second phase of HPβCD-induced cochlear damage. Thus, HPβCD-induced ototoxicity must be mediated by some other unknown cell-death pathway possibly involving loss of trophic support from damaged support cells or disrupted cholesterol metabolism.
Collapse
Affiliation(s)
- Senthilvelan Manohar
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, 14214, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Therapeutic Vision, Inc., Elkhorn, NE, 68022, USA
| | - Dalian Ding
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, 14214, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Therapeutic Vision, Inc., Elkhorn, NE, 68022, USA
| | - Haiyan Jiang
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, 14214, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Therapeutic Vision, Inc., Elkhorn, NE, 68022, USA
| | - Li Li
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, 14214, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Therapeutic Vision, Inc., Elkhorn, NE, 68022, USA
| | - Guang-Di Chen
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, 14214, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Therapeutic Vision, Inc., Elkhorn, NE, 68022, USA
| | - Peter Kador
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Therapeutic Vision, Inc., Elkhorn, NE, 68022, USA
| | - Richard Salvi
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, 14214, USA; Department of Pharmaceutical Sciences, College of Pharmacy, University of Nebraska Medical Center, Omaha, NE, 68198, USA; Therapeutic Vision, Inc., Elkhorn, NE, 68022, USA.
| |
Collapse
|
9
|
Chu TT, Tu X, Yang K, Wu J, Repa JJ, Yan N. Tonic prime-boost of STING signalling mediates Niemann-Pick disease type C. Nature 2021; 596:570-575. [PMID: 34290407 PMCID: PMC8859990 DOI: 10.1038/s41586-021-03762-2] [Citation(s) in RCA: 152] [Impact Index Per Article: 38.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/12/2020] [Accepted: 06/23/2021] [Indexed: 02/07/2023]
Abstract
The classic mode of STING activation is through binding the cyclic dinucleotide 2'3'-cyclic GMP-AMP (cGAMP), produced by the DNA sensor cyclic GMP-AMP synthase (cGAS), which is important for the innate immune response to microbial infection and autoimmune disease. Modes of STING activation that are independent of cGAS are much less well understood. Here, through a spatiotemporally resolved proximity labelling screen followed by quantitative proteomics, we identify the lysosomal membrane protein Niemann-Pick type C1 (NPC1) as a cofactor in the trafficking of STING. NPC1 interacts with STING and recruits it to the lysosome for degradation in both human and mouse cells. Notably, we find that knockout of Npc1 'primes' STING signalling by physically linking or 'tethering' STING to SREBP2 trafficking. Loss of NPC1 protein also 'boosts' STING signalling by blocking lysosomal degradation. Both priming and boosting of STING signalling are required for severe neurological disease in the Npc1-/- mouse. Genetic deletion of Sting1 (the gene that encodes STING) or Irf3, but not that of Cgas, significantly reduced the activation of microglia and relieved the loss of Purkinje neurons in the cerebellum of Npc1-/- mice, leading to improved motor function. Our study identifies a cGAS- and cGAMP-independent mode of STING activation that affects neuropathology and provides a therapeutic target for the treatment of Niemann-Pick disease type C.
Collapse
Affiliation(s)
- Ting-Ting Chu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Xintao Tu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Kun Yang
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Jianjun Wu
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joyce J Repa
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Nan Yan
- Department of Immunology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
- Department of Microbiology, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
10
|
Rodriguez-Gil JL, Baxter LL, Watkins-Chow DE, Johnson NL, Davidson CD, Carlson SR, Incao AA, Wallom KL, Farhat NY, Platt FM, Dale RK, Porter FD, Pavan WJ. Transcriptome of HPβCD-treated Niemann-pick disease type C1 cells highlights GPNMB as a biomarker for therapeutics. Hum Mol Genet 2021; 30:2456-2468. [PMID: 34296265 DOI: 10.1093/hmg/ddab194] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/17/2021] [Revised: 05/20/2021] [Accepted: 06/29/2021] [Indexed: 11/12/2022] Open
Abstract
The rare, fatal neurodegenerative disorder Niemann-Pick disease type C1 (NPC1) arises from lysosomal accumulation of unesterified cholesterol and glycosphingolipids. These subcellular pathologies lead to phenotypes of hepatosplenomegaly, neurological degeneration and premature death. The timing and severity of NPC1 clinical presentation is extremely heterogeneous. This study analyzed RNA-Seq data from 42 NPC1 patient-derived, primary fibroblast cell lines to determine transcriptional changes induced by treatment with 2-hydroxypropyl-β-cyclodextrin (HPβCD), a compound currently under investigation in clinical trials. A total of 485 HPβCD-responsive genes were identified. Pathway enrichment analysis of these genes showed significant involvement in cholesterol and lipid biosynthesis. Furthermore, immunohistochemistry of the cerebellum as well as measurements of serum from Npc1m1N null mice treated with HPβCD and adeno-associated virus (AAV) gene therapy suggests that one of the identified genes, GPNMB, may serve as a useful biomarker of treatment response in NPC1 disease. Overall, this large NPC1 patient-derived dataset provides a comprehensive foundation for understanding the genomic response to HPβCD treatment.
Collapse
Affiliation(s)
- Jorge L Rodriguez-Gil
- Genomics, Development and Disease Section, Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health.,Medical Scientist Training Program, University of Wisconsin-Madison School of Medicine and Public Health
| | - Laura L Baxter
- Genomics, Development and Disease Section, Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health
| | - Dawn E Watkins-Chow
- Genomics, Development and Disease Section, Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health
| | - Nicholas L Johnson
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health
| | - Cristin D Davidson
- Genomics, Development and Disease Section, Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health
| | - Steven R Carlson
- Genomics, Development and Disease Section, Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health
| | - Arturo A Incao
- Genomics, Development and Disease Section, Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health
| | | | | | - Nicole Y Farhat
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health
| | | | - Ryan K Dale
- Bioinformatics and Scientific Programming Core, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health
| | - Forbes D Porter
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health
| | - William J Pavan
- Genomics, Development and Disease Section, Genetic Disease Research Branch, National Human Genome Research Institute, National Institutes of Health
| |
Collapse
|
11
|
Cruz DL, Pipalia N, Mao S, Gadi D, Liu G, Grigalunas M, O'Neill M, Quinn TR, Kipper A, Ekebergh A, Dimmling A, Gartner C, Melancon BJ, Wagner FF, Holson E, Helquist P, Wiest O, Maxfield FR. Inhibition of Histone Deacetylases 1, 2, and 3 Enhances Clearance of Cholesterol Accumulation in Niemann-Pick C1 Fibroblasts. ACS Pharmacol Transl Sci 2021; 4:1136-1148. [PMID: 34151204 PMCID: PMC8204796 DOI: 10.1021/acsptsci.1c00033] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2021] [Indexed: 11/29/2022]
Abstract
![]()
Niemann-Pick disease type C1 (NPC1) is a rare genetic cholesterol storage disorder
caused by mutations in the NPC1 gene. Mutations in this transmembrane
late endosome protein lead to loss of normal cholesterol efflux from late endosomes and
lysosomes. It has been shown that broad spectrum histone deacetylase inhibitors
(HDACi's) such as Vorinostat correct the cholesterol accumulation phenotype in the
majority of NPC1 mutants tested in cultured cells. In order to determine the optimal
specificity for HDACi correction of the mutant NPC1s, we screened 76 HDACi's of varying
specificity. We tested the ability of these HDACi's to correct the excess accumulation
of cholesterol in patient fibroblast cells that homozygously express
NPC1I1061T, the most common mutation. We
determined that inhibition of HDACs 1, 2, and 3 is important for correcting the defect,
and combined inhibition of all three is needed to achieve the greatest effect,
suggesting a need for multiple effects of the HDACi treatments. Identifying the specific
HDACs involved in the process of regulating cholesterol trafficking in NPC1 will help to
focus the search for more specific druggable targets.
Collapse
Affiliation(s)
- Dana L Cruz
- Department of Biochemistry, Weill Cornell Medical College, New York, New York 10065, United States
| | - Nina Pipalia
- Department of Biochemistry, Weill Cornell Medical College, New York, New York 10065, United States
| | - Shu Mao
- Department of Biochemistry, Weill Cornell Medical College, New York, New York 10065, United States
| | - Deepti Gadi
- Department of Biochemistry, Weill Cornell Medical College, New York, New York 10065, United States
| | - Gang Liu
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Michael Grigalunas
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Matthew O'Neill
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Taylor R Quinn
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Andi Kipper
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Andreas Ekebergh
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Alexander Dimmling
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Carlos Gartner
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Bruce J Melancon
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Florence F Wagner
- Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States
| | - Edward Holson
- Eli and Edythe L. Broad Institute of MIT and Harvard, Cambridge, Massachusetts 02142, United States.,KDAc Therapeutics, Cambridge, Massachusetts 02142, United States
| | - Paul Helquist
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States
| | - Olaf Wiest
- Department of Chemistry and Biochemistry, University of Notre Dame, Notre Dame, Indiana 46556, United States.,Laboratory of Computational Chemistry and Drug Design, State Key Laboratory of Chemical Oncogenomics, Peking University, Shenzhen Graduate School, Shenzhen 518055, P.R. China
| | - Frederick R Maxfield
- Department of Biochemistry, Weill Cornell Medical College, New York, New York 10065, United States
| |
Collapse
|
12
|
Ding D, Jiang H, Manohar S, Liu X, Li L, Chen GD, Salvi R. Spatiotemporal Developmental Upregulation of Prestin Correlates With the Severity and Location of Cyclodextrin-Induced Outer Hair Cell Loss and Hearing Loss. Front Cell Dev Biol 2021; 9:643709. [PMID: 34109172 PMCID: PMC8181405 DOI: 10.3389/fcell.2021.643709] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Accepted: 03/08/2021] [Indexed: 11/24/2022] Open
Abstract
2-Hyroxypropyl-beta-cyclodextrin (HPβCD) is being used to treat Niemann-Pick C1, a fatal neurodegenerative disease caused by abnormal cholesterol metabolism. HPβCD slows disease progression, but unfortunately causes severe, rapid onset hearing loss by destroying the outer hair cells (OHC). HPβCD-induced damage is believed to be related to the expression of prestin in OHCs. Because prestin is postnatally upregulated from the cochlear base toward the apex, we hypothesized that HPβCD ototoxicity would spread from the high-frequency base toward the low-frequency apex of the cochlea. Consistent with this hypothesis, cochlear hearing impairments and OHC loss rapidly spread from the high-frequency base toward the low-frequency apex of the cochlea when HPβCD administration shifted from postnatal day 3 (P3) to P28. HPβCD-induced histopathologies were initially confined to the OHCs, but between 4- and 6-weeks post-treatment, there was an unexpected, rapid and massive expansion of the lesion to include most inner hair cells (IHC), pillar cells (PC), peripheral auditory nerve fibers, and spiral ganglion neurons at location where OHCs were missing. The magnitude and spatial extent of HPβCD-induced OHC death was tightly correlated with the postnatal day when HPβCD was administered which coincided with the spatiotemporal upregulation of prestin in OHCs. A second, massive wave of degeneration involving IHCs, PC, auditory nerve fibers and spiral ganglion neurons abruptly emerged 4–6 weeks post-HPβCD treatment. This secondary wave of degeneration combined with the initial OHC loss results in a profound, irreversible hearing loss.
Collapse
Affiliation(s)
- Dalian Ding
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, United States
| | - Haiyan Jiang
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, United States
| | - Senthilvelan Manohar
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, United States
| | - Xiaopeng Liu
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, United States
| | - Li Li
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, United States
| | - Guang-Di Chen
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, United States
| | - Richard Salvi
- Center for Hearing and Deafness, University at Buffalo, Buffalo, NY, United States
| |
Collapse
|
13
|
Yamada Y, Ishitsuka Y, Kondo Y, Nakahara S, Nishiyama A, Takeo T, Nakagata N, Motoyama K, Higashi T, Arima H, Kamei S, Shuto T, Kai H, Hayashino Y, Sugita M, Kikuchi T, Hirata F, Miwa T, Takeda H, Orita Y, Seki T, Ohta T, Kurauchi Y, Katsuki H, Matsuo M, Higaki K, Ohno K, Matsumoto S, Era T, Irie T. Differential mode of cholesterol inclusion with 2-hydroxypropyl-cyclodextrins increases safety margin in treatment of Niemann-Pick disease type C. Br J Pharmacol 2021; 178:2727-2746. [PMID: 33782944 DOI: 10.1111/bph.15464] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2020] [Revised: 03/05/2021] [Accepted: 03/10/2021] [Indexed: 11/29/2022] Open
Abstract
BACKGROUND AND PURPOSE Niemann-Pick disease type C (NPC) is a lysosomal storage disorder with disrupted intracellular cholesterol trafficking. A cyclic heptasaccharide, 2-hydroxypropyl-β-cyclodextrin (HP-β-CD), is a cholesterol solubilizer that is being developed to treat NPC, but its ototoxicity and pulmonary toxicity remain important issues. We have characterized 2-hydroxypropyl-γ-cyclodextrin (HP-γ-CD), a cyclic octasaccharide with a larger cavity than HP-β-CD, as a candidate drug to treat NPC. However, the molecular target of HP-γ-CD with respect to NPC and its potential for clinical application are still unclear. EXPERIMENTAL APPROACH We investigated the mode of interaction between HP-γ-CD and cholesterol by phase-solubility analysis, proton NMR spectroscopy and molecular dynamics simulations. We then evaluated the therapeutic effects of HP-γ-CD compared with HP-β-CD using cellular and murine NPC models. Mouse auditory and pulmonary function tests were also conducted. KEY RESULTS HP-γ-CD solely formed a 1:1 inclusion complex with cholesterol with an affinity similar to that of HP-β-CD. In vitro, HP-γ-CD and HP-β-CD amelioration of NPC-related manifestations was almost equivalent at lower concentrations. However, at higher concentrations, the cholesterol inclusion mode of HP-β-CD shifted to the highly soluble 2:1 complex whereas that of HP-γ-CD maintained solely the 1:1 complex. The constant lower cholesterol solubilizing ability of HP-γ-CD conferred it with significantly reduced toxicity compared with HP-β-CD, but equal efficacy in treating a mouse model of NPC. CONCLUSIONS AND IMPLICATIONS HP-γ-CD can serve as a fine-tuned cholesterol solubilizer for the treatment of NPC with a wider safety margin than HP-β-CD in terms of ototoxicity and pulmonary toxicity.
Collapse
Affiliation(s)
- Yusei Yamada
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoichi Ishitsuka
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuki Kondo
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Shuichi Nakahara
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Asami Nishiyama
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Toru Takeo
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Kumamoto University, Kumamoto, Japan
| | - Naomi Nakagata
- Division of Reproductive Engineering, Center for Animal Resources and Development (CARD), Kumamoto University, Kumamoto, Japan
| | - Keiichi Motoyama
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Taishi Higashi
- Department of Physical Pharmaceutics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hidetoshi Arima
- Laboratory of Evidence-Based Pharmacotherapy, Daiichi University of Pharmacy, Fukuoka, Japan
| | - Shunsuke Kamei
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
| | - Tsuyoshi Shuto
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
| | - Hirofumi Kai
- Department of Molecular Medicine, Graduate School of Pharmaceutical Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto, Japan
| | - Yuji Hayashino
- Department of Bioinformatics, College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Masatake Sugita
- Department of Bioinformatics, College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Takeshi Kikuchi
- Department of Bioinformatics, College of Life Sciences, Ritsumeikan University, Kusatsu, Shiga, Japan
| | - Fumio Hirata
- Toyota Physical and Chemical Research Institute, Nagakute, Aichi, Japan
| | - Toru Miwa
- Department of Otolaryngology-Head and Neck Surgery, Tazuke Kofukai Medical Research Institute, Kitano Hospital, Osaka, Japan
| | - Hiroki Takeda
- Department of Otolaryngology-Head and Neck Surgery, Kumamoto University, Kumamoto, Japan
| | - Yorihisa Orita
- Department of Otolaryngology-Head and Neck Surgery, Kumamoto University, Kumamoto, Japan
| | - Takahiro Seki
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tomoko Ohta
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuki Kurauchi
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Hiroshi Katsuki
- Department of Chemico-Pharmacological Sciences, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Muneaki Matsuo
- Department of Pediatrics, Faculty of Medicine, Saga University, Saga, Japan
| | - Katsumi Higaki
- Research Initiative Center, Organization for Research Initiative and Promotion, Tottori University, Yonago, Japan
| | | | - Shirou Matsumoto
- Department of Pediatrics, Graduate School of Medical Sciences, Kumamoto University, Kumamoto, Japan
| | - Takumi Era
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Tetsumi Irie
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| |
Collapse
|
14
|
Ramirez CM, Taylor AM, Lopez AM, Repa JJ, Turley SD. Delineation of metabolic responses of Npc1 -/-nih mice lacking the cholesterol-esterifying enzyme SOAT2 to acute treatment with 2-hydroxypropyl-β-cyclodextrin. Steroids 2020; 164:108725. [PMID: 32890578 PMCID: PMC7680374 DOI: 10.1016/j.steroids.2020.108725] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/08/2020] [Revised: 08/14/2020] [Accepted: 08/25/2020] [Indexed: 12/26/2022]
Abstract
Lipids present in lipoproteins cleared from the circulation are processed sequentially by three major proteins within the late endosomal/lysosomal (E/L) compartment of all cells: lysosomal acid lipase (LAL), Niemann-Pick (NPC) C2 and NPC1. When all three of these proteins are functioning normally, unesterified cholesterol (UC) exits the E/L compartment and is used in plasma membrane maintenance and various pathways in the endoplasmic reticulum including esterification by sterol O-acyltransferase 2 (SOAT2) or SOAT1 depending partly on cell type. Mutations in either NPC2 or NPC1 result in continual entrapment of UC and glycosphingolipids leading to neurodegeneration, pulmonary dysfunction, splenomegaly and liver damage. To date, the most effective agent for promoting release of entrapped UC in nearly all organs of NPC1-deficient mice and cats is 2-hydroxypropyl-β-cyclodextrin (2HPβCD). The cytotoxic nature of the liberated UC triggers various defenses including suppression of sterol synthesis and increased esterification. The present studies, using the Npc1-/-nih mouse model, measured the comparative quantitative importance of these two responses in the liver versus the spleen of Npc1-/-: Soat2+/+ and Npc1-/-: Soat2-/- mice in the 24 h following a single acute treatment with 2HPβCD. In the liver but not the spleen of both types of mice suppression of synthesis alone or in combination with increased esterification provided the major defense against the rise in unsequestered cellular UC content. These findings have implications for systemic 2HPβCD treatment in NPC1 patients in view of the purportedly low levels of SOAT2 activity in human liver.
Collapse
Affiliation(s)
- Charina M Ramirez
- Department of Pediatrics, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Anna M Taylor
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Adam M Lopez
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Joyce J Repa
- Department of Physiology, University of Texas Southwestern Medical Center, Dallas, TX, USA; Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | - Stephen D Turley
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA.
| |
Collapse
|
15
|
Ding D, Jiang H, Salvi R. Cochlear spiral ganglion neuron degeneration following cyclodextrin-induced hearing loss. Hear Res 2020; 400:108125. [PMID: 33302057 DOI: 10.1016/j.heares.2020.108125] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/08/2020] [Revised: 11/10/2020] [Accepted: 11/19/2020] [Indexed: 01/12/2023]
Abstract
Because cyclodextrins are capable of removing cholesterol from cell membranes, there is growing interest in using these compounds to treat diseases linked to aberrant cholesterol metabolism. One compound, 2-hydroxypropyl-beta-cyclodextrin (HPβCD), is currently being evaluated as a treatment for Niemann-Pick Type C1 disease, a rare, fatal neurodegenerative disease caused by the buildup of lipids in endosomes and lysosomes. HPβCD can reduce some debilitating symptoms and extend life span, but the therapeutic doses used to treat the disease cause hearing loss. Initial studies in rodents suggested that HPβCD selectively damaged only cochlear outer hair cells during the first week post-treatment. However, our recent in vivo and in vitro studies suggested that the damage could become progressively worse and more extensive over time. To test this hypothesis, we treated rats subcutaneously with 1, 2, 3 or 4 g/kg of HPβCD and waited for 8-weeks to assess the long-term histological consequences. Our new results indicate that the two highest doses of HPβCD caused extensive damage not only to OHC, but also to inner hair cells, pillar cells and other support cells resulting in the collapse and flattening of the sensory epithelium. The 4 g/kg dose destroyed all the outer hair cells and three-fourths of the inner hair cells over the basal two-thirds of the cochlea and more than 85% of the nerve fibers in the habenula perforata and more than 80% of spiral ganglion neurons in the middle of basal turn of the cochlea. The mechanisms that lead to the delayed degeneration of inner hair cells, pillar cells, nerve fibers and spiral ganglion neurons remain poorly understood, but may be related to the loss of trophic support caused by the degeneration of sensory and/or support cells in the organ of Corti. Despite the massive damage to the cochlear sensory epithelium, the blood vessels in the stria vascularis and the vestibular hair cells in the utricle and saccule remained normal.
Collapse
Affiliation(s)
- Dalian Ding
- Center for Hearing and Deafness, University at Buffalo, 137 Cary Hall, Buffalo, NY 14221, USA
| | - Haiyan Jiang
- Center for Hearing and Deafness, University at Buffalo, 137 Cary Hall, Buffalo, NY 14221, USA
| | - Richard Salvi
- Center for Hearing and Deafness, University at Buffalo, 137 Cary Hall, Buffalo, NY 14221, USA.
| |
Collapse
|
16
|
Subramanian K, Hutt DM, Scott SM, Gupta V, Mao S, Balch WE. Correction of Niemann-Pick type C1 trafficking and activity with the histone deacetylase inhibitor valproic acid. J Biol Chem 2020; 295:8017-8035. [PMID: 32354745 DOI: 10.1074/jbc.ra119.010524] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2019] [Revised: 04/13/2020] [Indexed: 12/12/2022] Open
Abstract
Niemann-Pick type C (NPC) disease is primarily caused by mutations in the NPC1 gene and is characterized by the accumulation of unesterified cholesterol and lipids in the late endosomal (LE) and lysosomal (Ly) compartments. The most prevalent disease-linked mutation is the I1061T variant of NPC1, which exhibits defective folding and trafficking from the endoplasmic reticulum to the LE/Ly compartments. We now show that the FDA-approved histone deacetylase inhibitor (HDACi) valproic acid (VPA) corrects the folding and trafficking defect associated with I1061T-NPC1 leading to restoration of cholesterol homeostasis, an effect that is largely driven by a reduction in HDAC7 expression. The VPA-mediated trafficking correction is in part associated with an increase in the acetylation of lysine residues in the cysteine-rich domain of NPC1. The HDACi-mediated correction is synergistically improved by combining it with the FDA-approved anti-malarial, chloroquine, a known lysosomotropic compound, which improved the stability of the LE/Ly-localized fraction of the I1061T variant. We posit that combining the activity of VPA, to modulate epigenetically the cellular acetylome, with chloroquine, to alter the lysosomal environment to favor stability of the trafficked I1061T variant protein can have a significant therapeutic benefit in patients carrying at least one copy of the I1061T variant of NPC1, the most common disease-associated mutation leading to NPC disease. Given its ability to cross the blood-brain barrier, we posit VPA provides a potential mechanism to improve the response to 2-hydroxypropyl-β-cyclodextrin, by restoring a functional NPC1 to the cholesterol managing compartment as an adjunct therapy.
Collapse
Affiliation(s)
| | - Darren M Hutt
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Samantha M Scott
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Vijay Gupta
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| | - Shu Mao
- Department of Biochemistry, Weill Cornell Medical College, New York, New York, USA
| | - William E Balch
- Department of Molecular Medicine, Scripps Research, La Jolla, California, USA
| |
Collapse
|
17
|
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
|
18
|
Feltes M, Gale SE, Moores S, Ory DS, Schaffer JE. Monitoring the itinerary of lysosomal cholesterol in Niemann-Pick Type C1-deficient cells after cyclodextrin treatment. J Lipid Res 2020; 61:403-412. [PMID: 31988149 DOI: 10.1194/jlr.ra119000571] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 01/16/2020] [Indexed: 11/20/2022] Open
Abstract
Niemann-Pick disease type C (NPC) disease is a lipid-storage disorder that is caused by mutations in the genes encoding NPC proteins and results in lysosomal cholesterol accumulation. 2-Hydroxypropyl-β-cyclodextrin (CD) has been shown to reduce lysosomal cholesterol levels and enhance sterol homeostatic responses, but CD's mechanism of action remains unknown. Recent work provides evidence that CD stimulates lysosomal exocytosis, raising the possibility that lysosomal cholesterol is released in exosomes. However, therapeutic concentrations of CD do not alter total cellular cholesterol, and cholesterol homeostatic responses at the ER are most consistent with increased ER membrane cholesterol. To address these disparate findings, here we used stable isotope labeling to track the movement of lipoprotein cholesterol cargo in response to CD in NPC1-deficient U2OS cells. Although released cholesterol was detectable, it was not associated with extracellular vesicles. Rather, we demonstrate that lysosomal cholesterol trafficks to the plasma membrane (PM), where it exchanges with lipoprotein-bound cholesterol in a CD-dependent manner. We found that in the absence of suitable extracellular cholesterol acceptors, cholesterol exchange is abrogated, cholesterol accumulates in the PM, and reesterification at the ER is increased. These results support a model in which CD promotes intracellular redistribution of lysosomal cholesterol, but not cholesterol exocytosis or efflux, during the restoration of cholesterol homeostatic responses.
Collapse
Affiliation(s)
- McKenna Feltes
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Sarah E Gale
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Samantha Moores
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Daniel S Ory
- Department of Medicine, Washington University School of Medicine, St. Louis, MO
| | - Jean E Schaffer
- Department of Medicine, Washington University School of Medicine, St. Louis, MO .,Joslin Diabetes Center, Harvard Medical School, Boston, MA
| |
Collapse
|
19
|
Lopez AM, Ramirez CM, Taylor AM, Jones RD, Repa JJ, Turley SD. Ontogenesis and Modulation of Intestinal Unesterified Cholesterol Sequestration in a Mouse Model of Niemann-Pick C1 Disease. Dig Dis Sci 2020; 65:158-167. [PMID: 31312996 DOI: 10.1007/s10620-019-05736-x] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2019] [Accepted: 07/11/2019] [Indexed: 01/24/2023]
Abstract
BACKGROUND Mutations in the NPC1 gene result in sequestration of unesterified cholesterol (UC) and glycosphingolipids in most tissues leading to multi-organ disease, especially in the brain, liver, lungs, and spleen. Various data from NPC1-deficient mice suggest the small intestine (SI) is comparatively less affected, even in late stage disease. METHODS Using the Npc1nih mouse model, we measured SI weights and total cholesterol (TC) levels in Npc1-/- versus Npc1+/+ mice as a function of age, and then after prolonged ezetimibe-induced inhibition of cholesterol absorption. Next, we determined intestinal levels of UC and esterified cholesterol (EC), and cholesterol synthesis rates in Npc1-/- and Npc1+/+ mice, with and without the cholesterol-esterifying enzyme SOAT2, following a once-only subcutaneous injection with 2-hydroxypropyl-β-cyclodextrin (2HPβCD). RESULTS By ~ 42 days of age, intestinal TC levels averaged ~ 2.1-fold more (mostly UC) in the Npc1-/- versus Npc1+/+ mice with no further increase thereafter. Chronic ezetimibe treatment lowered intestinal TC levels in the Npc1-/- mice by only ~ 16%. In Npc1-/- mice given 2HPβCD 24 h earlier, UC levels fell, EC levels increased (although less so in mice lacking SOAT2), and cholesterol synthesis was suppressed equally in the Npc1-/-:Soat2+/+ and Npc1-/-:Soat2-/- mice. CONCLUSIONS The low and static levels of intestinal UC sequestration in Npc1-/- mice likely reflect the continual sloughing of cells from the mucosa. This sequestration is blunted by about the same extent following a single acute treatment with 2HPβCD as it is by a prolonged ezetimibe-induced block of cholesterol absorption.
Collapse
MESH Headings
- 2-Hydroxypropyl-beta-cyclodextrin/pharmacology
- Animals
- Cholesterol/metabolism
- Disease Models, Animal
- Ezetimibe/pharmacology
- Female
- Intestinal Absorption/drug effects
- Intestinal Mucosa/drug effects
- Intestinal Mucosa/metabolism
- Intestine, Small/drug effects
- Intestine, Small/metabolism
- Intracellular Signaling Peptides and Proteins/deficiency
- Intracellular Signaling Peptides and Proteins/genetics
- Male
- Mice, 129 Strain
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Niemann-Pick C1 Protein
- Niemann-Pick Disease, Type C/drug therapy
- Niemann-Pick Disease, Type C/genetics
- Niemann-Pick Disease, Type C/metabolism
- Sterol O-Acyltransferase/genetics
- Sterol O-Acyltransferase/metabolism
- Sterol O-Acyltransferase 2
Collapse
Affiliation(s)
- Adam M Lopez
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Charina M Ramirez
- Department of Pediatrics, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Anna M Taylor
- Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
- School of Behavioral and Brain Sciences, University of Texas at Dallas, Richardson, TX, 75080, USA
| | - Ryan D Jones
- Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
- Department of Pathology, Northwestern University, Chicago, IL, 60611, USA
| | - Joyce J Repa
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
- Department of Physiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA
| | - Stephen D Turley
- Department of Internal Medicine, University of Texas Southwestern Medical Center, 5323 Harry Hines Blvd, Dallas, TX, 75390, USA.
| |
Collapse
|
20
|
Hastings C, Vieira C, Liu B, Bascon C, Gao C, Wang RY, Casey A, Hrynkow S. Expanded access with intravenous hydroxypropyl-β-cyclodextrin to treat children and young adults with Niemann-Pick disease type C1: a case report analysis. Orphanet J Rare Dis 2019; 14:228. [PMID: 31639011 PMCID: PMC6805667 DOI: 10.1186/s13023-019-1207-1] [Citation(s) in RCA: 27] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2019] [Accepted: 09/21/2019] [Indexed: 12/15/2022] Open
Abstract
Background Niemann-Pick Disease Type C (NPC) is an inherited, often fatal neurovisceral lysosomal storage disease characterized by cholesterol accumulation in every cell with few known treatments. Defects in cholesterol transport cause sequestration of unesterified cholesterol within the endolysosomal system. The discovery that systemic administration of hydroxypropyl-beta cyclodextrin (HPβPD) to NPC mice could release trapped cholesterol from lysosomes, normalize cholesterol levels in the liver, and prolong life, led to expanded access use in NPC patients. HPβCD has been administered to NPC patients with approved INDs globally since 2009. Results Here we present safety, tolerability and efficacy data from 12 patients treated intravenously (IV) for over 7 years with HPβCD in the US and Brazil. Some patients subsequently received intrathecal (IT) treatment with HPβCD following on average 13 months of IV HPβCD. Several patients transitioned to an alternate HPβCD. Moderately affected NPC patients treated with HPβCD showed slowing of disease progression. Severely affected patients demonstrated periods of stability but eventually showed progression of disease. Neurologic and neurocognitive benefits were seen in most patients with IV alone, independent of the addition of IT administration. Physicians and caregivers reported improvements in quality of life for the patients on IV therapy. There were no safety issues, and the drug was well tolerated and easy to administer. Conclusions These expanded access data support the safety and potential benefit of systemic IV administration of HPβCD and provide a platform for two clinical trials to study the effect of intravenous administration of HPβCD in NPC patients.
Collapse
Affiliation(s)
- Caroline Hastings
- Department of Pediatric Hematology Oncology, UCSF Benioff Children's Hospital Oakland, 747 52nd Street, Oakland, CA, 94609-1809, USA. .,Department of Pediatrics, University of California San Francisco, San Francisco, CA, USA.
| | - Camilo Vieira
- Universidade Federal da Bahia, Clínica Citta, Ed. Mundo Plaza, Av. Tancredo Neves, 620, Sala 1905, Camino dos Árvares, Salvador, Brazil
| | - Benny Liu
- GI & Liver Clinics, Highland Hospital, Alameda Health System, Highland Hospital, Oakland, CA, USA.,Division of Gastroenterology & Hepatology, Highland Hospital, Alameda Health Systems, Highland Care Pavilion 5th floor, 1411 East 31st Street, Oakland, CA, 94602, USA
| | - Cyrus Bascon
- Department of Pediatric Hematology Oncology, UCSF Benioff Children's Hospital Oakland, 747 52nd Street, Oakland, CA, 94609-1809, USA
| | - Claire Gao
- UCSF Benioff Children's Hospital Oakland, Oakland, CA, USA.,Present Address: Neuroscience Graduate Program, Brown University, 185 Meeting Street, Box GL-N, Providence, RI, 02912, USA
| | - Raymond Y Wang
- Division of Metabolic Disorders, Children's Hospital of Orange County, CHOC Children's Specialists, 1201 W. La Veta Ave, Orange, CA, 92868, USA.,Department of Pediatrics, University of California, Irvine School of Medicine, Irvine, CA, 92868, USA
| | - Alicia Casey
- Boston Children's Hospital, 300 Longwood Avenue, Boston, MA, 02115, USA
| | - Sharon Hrynkow
- CTD Holdings, Inc., P.O. Box 1180, Alachua, FL, 32616, USA
| |
Collapse
|
21
|
Neßlauer AM, Gläser A, Gräler M, Engelmann R, Müller-Hilke B, Frank M, Burstein C, Rolfs A, Neidhardt J, Wree A, Witt M, Bräuer AU. A therapy with miglustat, 2-hydroxypropyl-ß-cyclodextrin and allopregnanolone restores splenic cholesterol homeostasis in Niemann-pick disease type C1. Lipids Health Dis 2019; 18:146. [PMID: 31248418 PMCID: PMC6598286 DOI: 10.1186/s12944-019-1088-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2019] [Accepted: 06/11/2019] [Indexed: 01/02/2023] Open
Abstract
BACKGROUND Niemann-Pick disease type C1 (NPC1) is an autosomal-recessive lipid-storage disorder with an estimated minimal incidence of 1/120,000 live births. Besides other neuronal and visceral symptoms, NPC1 patients develop spleen dysfunction, isolated spleno- or hepatosplenomegaly and infections. The mechanisms of splenomegaly and alterations of lipid metabolism-related genes in NPC1 disease are still poorly understood. METHODS Here, we used an NPC1 mouse model to study a splenoprotective effect of a treatment with miglustat, 2-hydroxypropyl-ß-cyclodextrin and allopregnanolone and showed that this treatment has a positive effect on spleen morphology and lipid metabolism. RESULTS Disease progress can be halted and blocked at the molecular level. Mutant Npc1 (Npc1-/-) mice showed increased spleen weight and increased lipid accumulation that could be avoided by our treatment. Also, FACS analyses showed that the increased number of splenic myeloid cells in Npc1-/- mice was normalized by the treatment. Treated Npc1-/- mice showed decreased numbers of cytotoxic T cells and increased numbers of T helper cells. CONCLUSIONS In summary, the treatment promotes normal spleen morphology, stabilization of lipid homeostasis and blocking of inflammation, but alters the composition of T cell subtypes.
Collapse
Affiliation(s)
- Anna-Maria Neßlauer
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstraße 9, 18057, Rostock, Germany
| | - Anne Gläser
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstraße 9, 18057, Rostock, Germany.,Research Group Anatomy, School of Medicine and Health Sciences, Department für Humanmedizin, Abteilung Anatomie, Carl von Ossietzky University Oldenburg, Carl-von-Ossietzky Straße 9-11, 26129, Oldenburg, Germany
| | - Markus Gräler
- Department of Anesthesiology and Intensive Care Medicine, Center for Sepsis Control and Care (CSCC), and the Center for Molecular Biomedicine (CMB), Jena University Hospital, Hans-Knöll-Str. 2, 07745, Jena, Germany
| | - Robby Engelmann
- Institute of Immunology, Rostock University Medical Center, Schillingallee 70, 18057, Rostock, Germany
| | - Brigitte Müller-Hilke
- Institute of Immunology, Rostock University Medical Center, Schillingallee 70, 18057, Rostock, Germany
| | - Marcus Frank
- Medical Biology and Electron Microscopy Center, Rostock University Medical Center, Strempelstraße 14, 18057, Rostock, Germany
| | - Christine Burstein
- Institute of Clinical Chemistry and Pathobiochemistry, Rostock University Medical Center, Ernst-Heydemann-Straße 6, 18057, Rostock, Germany
| | - Arndt Rolfs
- Centogene AG, Am Strande 7, 18055, Rostock, Germany
| | - John Neidhardt
- Human Genetics, Faculty of Medicine and Health Sciences, University of Oldenburg, Oldenburg, Germany.,Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany
| | - Andreas Wree
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstraße 9, 18057, Rostock, Germany
| | - Martin Witt
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstraße 9, 18057, Rostock, Germany
| | - Anja U Bräuer
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstraße 9, 18057, Rostock, Germany. .,Research Group Anatomy, School of Medicine and Health Sciences, Department für Humanmedizin, Abteilung Anatomie, Carl von Ossietzky University Oldenburg, Carl-von-Ossietzky Straße 9-11, 26129, Oldenburg, Germany. .,Research Center for Neurosensory Science, Carl von Ossietzky University Oldenburg, Oldenburg, Germany.
| |
Collapse
|
22
|
Vectisol Formulation Enhances Solubility of Resveratrol and Brings Its Benefits to Kidney Transplantation in a Preclinical Porcine Model. Int J Mol Sci 2019; 20:ijms20092268. [PMID: 31071925 PMCID: PMC6540035 DOI: 10.3390/ijms20092268] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2019] [Revised: 05/03/2019] [Accepted: 05/05/2019] [Indexed: 02/07/2023] Open
Abstract
Current organ shortages have led centers to extend the acceptance criteria for organs, increasing the risk for adverse outcomes. Current preservation protocols have not been adapted so as to efficiently protect these organs. Herein, we target oxidative stress, the key mechanism of ischemia reperfusion injury. Vectisol® is a novel antioxidant strategy based on the encapsulation of resveratrol into a cyclodextrin, increasing its bioavailability. We tested this compound as an additive to the most popular static preservation solutions and machine perfusion (LifePort) in a preclinical pig model of kidney autotransplantation. In regard to static preservation, supplementation improved glomerular filtration and proximal tubular function early recovery. Extended follow-up confirmed the higher level of protection, slowing chronic loss of function (creatininemia and proteinuria) and the onset of histological lesions. Regarding machine perfusion, the use of Vectisol® decreased oxidative stress and apoptosis at the onset of reperfusion (30 min post declamping). Improved quality was confirmed with decreased early levels of circulating SOD (Superoxide Dismutase) and ASAT (asparagine amino transferase). Supplementation slowed the onset of chronic loss of function, as well as interstitial fibrosis and tubular atrophy. The simple addition of Vectisol® to the preservation solution significantly improved the performance of organ preservation, with long-term effects on the outcome. This strategy is thus a key player for future multi-drug therapy aimed at ischemia reperfusion in transplantation.
Collapse
|
23
|
Lopez AM, Jones RD, Repa JJ, Turley SD. Niemann-Pick C1-deficient mice lacking sterol O-acyltransferase 2 have less hepatic cholesterol entrapment and improved liver function. Am J Physiol Gastrointest Liver Physiol 2018; 315:G454-G463. [PMID: 29878847 PMCID: PMC6230690 DOI: 10.1152/ajpgi.00124.2018] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/11/2018] [Revised: 05/31/2018] [Accepted: 06/01/2018] [Indexed: 01/31/2023]
Abstract
Cholesteryl esters are generated at multiple sites in the body by sterol O-acyltransferase (SOAT) 1 or SOAT2 in various cell types and lecithin cholesterol acyltransferase in plasma. Esterified cholesterol and triacylglycerol contained in lipoproteins cleared from the circulation via receptor-mediated or bulk-phase endocytosis are hydrolyzed by lysosomal acid lipase within the late endosomal/lysosomal (E/L) compartment. Then, through the successive actions of Niemann-Pick C (NPC) 2 and NPC 1, unesterified cholesterol (UC) is exported from the E/L compartment to the cytosol. Mutations in either NPC1 or NPC2 lead to continuing entrapment of UC in all organs, resulting in multisystem disease, which includes hepatic dysfunction and in some cases liver failure. These studies investigated primarily whether elimination of SOAT2 in NPC1-deficient mice impacted hepatic UC sequestration, inflammation, and transaminase activities. Measurements were made in 7-wk-old mice fed a low-cholesterol chow diet or one enriched with cholesterol starting 2 wk before study. In the chow-fed mice, NPC1:SOAT2 double knockouts, compared with their littermates lacking only NPC1, had 20% less liver mass, 28% lower hepatic UC concentrations, and plasma alanine aminotransferase and aspartate aminotransferase activities that were decreased by 48% and 36%, respectively. mRNA expression levels for several markers of inflammation were all significantly lower in the NPC1 mutants lacking SOAT2. The existence of a new class of potent and selective SOAT2 inhibitors provides an opportunity for exploring if suppression of this enzyme could potentially become an adjunctive therapy for liver disease in NPC1 deficiency. NEW & NOTEWORTHY In Niemann-Pick type C1 (NPC1) disease, the entrapment of unesterified cholesterol (UC) in the endosomal/lysosomal compartment of all cells causes multiorgan disease, including neurodegeneration, pulmonary dysfunction, and liver failure. Some of this sequestered UC entered cells initially in the esterified form. When sterol O-acyltransferase 2, a cholesterol esterifying enzyme present in enterocytes and hepatocytes, is eliminated in NPC1-deficient mice, there is a reduction in their hepatomegaly, hepatic UC content, and cellular injury.
Collapse
Affiliation(s)
- Adam M Lopez
- Department of Internal Medicine, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Ryan D Jones
- Department of Physiology, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Joyce J Repa
- Department of Internal Medicine, University of Texas Southwestern Medical Center , Dallas, Texas
- Department of Physiology, University of Texas Southwestern Medical Center , Dallas, Texas
| | - Stephen D Turley
- Department of Internal Medicine, University of Texas Southwestern Medical Center , Dallas, Texas
| |
Collapse
|
24
|
Zhou Y, Takahashi S, Homma K, Duan C, Zheng J, Cheatham MA, Zheng J. The susceptibility of cochlear outer hair cells to cyclodextrin is not related to their electromotile activity. Acta Neuropathol Commun 2018; 6:98. [PMID: 30249300 PMCID: PMC6151916 DOI: 10.1186/s40478-018-0599-9] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2018] [Accepted: 09/13/2018] [Indexed: 11/26/2022] Open
Abstract
Niemann-Pick Type C1 (NPC1) disease is a fatal neurovisceral disorder caused by dysfunction of NPC1 protein, which plays a role in intracellular cholesterol trafficking. The cholesterol-chelating agent, 2-hydroxypropyl-β-cyclodextrin (HPβCD), is currently undergoing clinical trials for treatment of this disease. Though promising in alleviating neurological symptoms, HPβCD causes irreversible hearing loss in NPC1 patients and outer hair cell (OHC) death in animal models. We recently found that HPβCD-induced OHC death can be significantly alleviated in a mouse model lacking prestin, an OHC-specific motor protein required for the high sensitivity and sharp frequency selectivity of mammalian hearing. Since cholesterol status is known to influence prestin’s electromotility, we examined how prestin contributes to HPβCD-induced OHC death in the disease context using the NPC1 knockout (KO) mouse model (NPC1-KO). We found normal expression and localization of prestin in NPC1-KO OHCs. Whole-cell patch-clamp recordings revealed a significant depolarization of the voltage-operating point of prestin in NPC1-KO mice, suggesting reduced levels of cholesterol in the lateral membrane of OHCs that lack NPC1. OHC loss and elevated thresholds were found for high frequency regions in NPC1-KO mice, whose OHCs retained their sensitivity to HPβCD. To investigate whether prestin’s electromotile function contributes to HPβCD-induced OHC death, the prestin inhibitor salicylate was co-administered with HPβCD to WT and NPC1-KO mice. Neither oral nor intraperitoneal administration of salicylate mitigated HPβCD-induced OHC loss. To further determine the contribution of prestin’s electromotile function, a mouse model expressing a virtually nonelectromotile prestin protein (499-prestin) was subjected to HPβCD treatment. 499-prestin knockin mice showed no resistance to HPβCD-induced OHC loss. As 499-prestin maintains its ability to bind cholesterol, our data imply that HPβCD-induced OHC death is ascribed to the structural role of prestin in maintaining the OHC’s lateral membrane, rather than its motor function.
Collapse
|
25
|
Matencio A, Alcaráz-Gómez MA, García-Carmona F, Arias B, López-Nicolás JM. Application of a simple methodology to analyze Hydroxypropyl-β-Cyclodextrin in urine using HPLC–LS in early Niemann–Pick disease type C patient. J Chromatogr B Analyt Technol Biomed Life Sci 2018; 1093-1094:47-51. [DOI: 10.1016/j.jchromb.2018.06.051] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2018] [Revised: 06/19/2018] [Accepted: 06/23/2018] [Indexed: 11/25/2022]
|
26
|
Linear Cyclodextrin Polymer Prodrugs as Novel Therapeutics for Niemann-Pick Type C1 Disorder. Sci Rep 2018; 8:9547. [PMID: 29934581 PMCID: PMC6015065 DOI: 10.1038/s41598-018-27926-9] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 06/13/2018] [Indexed: 12/14/2022] Open
Abstract
Niemann-Pick Type C1 disorder (NPC) is a rare lysosomal storage disease characterized by the accumulation of cholesterol in lysosomes. NPC has no FDA approved treatments yet, however 2-hydroxypropyl-β-cyclodextrin (HPβCD) has shown efficacy for treating the disease in both mouse and feline NPC models and is currently being investigated in late stage clinical trials. Despite promising results, therapeutic use of HPβCD is limited by the need for high doses, ototoxicity and intrathecal administration. These limitations can be attributed to its poor pharmacokinetic profile. In the attempt to overcome these limitations, we have designed a β-cyclodextrin (βCD) based polymer prodrugs (ORX-301) for an enhanced pharmacokinetic and biodistribution profile, which in turn can potentially provide an improved efficacy at lower doses. We demonstrated that subcutaneously injected ORX-301 extended the mean lifespan of NPC mice at a dosage 5-fold lower (800 mg/kg, body weight) the HPβCD dose proven efficacious (4000 mg/kg). We also show that ORX-301 penetrates the blood brain barrier and counteracts neurological impairment. These properties represent a substantial improvement and appear to overcome major limitations of presently available βCD-based therapy, demonstrating that this novel prodrug is a valuable alternative/complement for existing therapies.
Collapse
|
27
|
Sima N, Li R, Huang W, Xu M, Beers J, Zou J, Titus S, Ottinger EA, Marugan JJ, Xie X, Zheng W. Neural stem cells for disease modeling and evaluation of therapeutics for infantile (CLN1/PPT1) and late infantile (CLN2/TPP1) neuronal ceroid lipofuscinoses. Orphanet J Rare Dis 2018; 13:54. [PMID: 29631617 PMCID: PMC5891977 DOI: 10.1186/s13023-018-0798-2] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2017] [Accepted: 03/29/2018] [Indexed: 02/06/2023] Open
Abstract
BACKGROUND Infantile and late infantile neuronal ceroid lipofuscinoses (NCLs) are lysosomal storage diseases affecting the central nervous system (CNS). The infantile NCL (INCL) is caused by mutations in the PPT1 gene and late-infantile NCL (LINCL) is due to mutations in the TPP1 gene. Deficiency in PPT1 or TPP1 enzyme function results in lysosomal accumulation of pathological lipofuscin-like material in the patient cells. There is currently no small-molecular drug treatment for NCLs. RESULTS We have generated induced pluripotent stem cells (iPSC) from three patient dermal fibroblast lines and further differentiated them into neural stem cells (NSCs). Using these new disease models, we evaluated the effect of δ-tocopherol (DT) and hydroxypropyl-β-cyclodextrin (HPBCD) with the enzyme replacement therapy as the control. Treatment with the relevant recombinant enzyme or DT significantly ameliorated the lipid accumulation and lysosomal enlargement in the disease cells. A combination therapy of δ-tocopherol and HPBCD further improved the effect compared to that of either drug used as a single therapy. CONCLUSION The results demonstrate that these patient iPSC derived NCL NSCs are valid cell- based disease models with characteristic disease phenotypes that can be used for study of disease pathophysiology and drug development.
Collapse
Affiliation(s)
- Ni Sima
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD, 20892, USA.,Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Rong Li
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD, 20892, USA
| | - Wei Huang
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD, 20892, USA.,Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Miao Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD, 20892, USA
| | - Jeanette Beers
- iPSC core, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Jizhong Zou
- iPSC core, National Heart, Lung and Blood Institute, National Institutes of Health, Bethesda, MD, USA
| | - Steven Titus
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD, 20892, USA
| | - Elizabeth A Ottinger
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD, 20892, USA
| | - Juan J Marugan
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD, 20892, USA
| | - Xing Xie
- Department of Gynecologic Oncology, Women's Reproductive Health Laboratory of Zhejiang Province, Women's Hospital, School of Medicine, Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, 9800 Medical Center Drive, Bethesda, MD, 20892, USA.
| |
Collapse
|
28
|
Abstract
Bietti’s crystalline dystrophy (BCD) is an autosomal recessive, progressive chorioretinal degenerative disease. Retinal pigment epithelium (RPE) cells are impaired in patients with BCD, but the underlying mechanisms of RPE cell damage have not yet been determined because cells from lesions cannot be readily acquired from patients with BCD. In the present study, we successfully generated a human in vitro model of BCD, BCD patient-specific iPSC-RPE cells, and demonstrated that the accumulation of free cholesterol caused RPE cell damage and subsequent cell death via the induction of lysosomal dysfunction and impairment of autophagy flux in BCD-affected cells. We believe these findings provide evidence of the possible therapeutic efficacy of reducing intracellular free cholesterol in BCD. Bietti’s crystalline dystrophy (BCD) is an intractable and progressive chorioretinal degenerative disease caused by mutations in the CYP4V2 gene, resulting in blindness in most patients. Although we and others have shown that retinal pigment epithelium (RPE) cells are primarily impaired in patients with BCD, the underlying mechanisms of RPE cell damage are still unclear because we lack access to appropriate disease models and to lesion-affected cells from patients with BCD. Here, we generated human RPE cells from induced pluripotent stem cells (iPSCs) derived from patients with BCD carrying a CYP4V2 mutation and successfully established an in vitro model of BCD, i.e., BCD patient-specific iPSC-RPE cells. In this model, RPE cells showed degenerative changes of vacuolated cytoplasm similar to those in postmortem specimens from patients with BCD. BCD iPSC-RPE cells exhibited lysosomal dysfunction and impairment of autophagy flux, followed by cell death. Lipidomic analyses revealed the accumulation of glucosylceramide and free cholesterol in BCD-affected cells. Notably, we found that reducing free cholesterol by cyclodextrins or δ-tocopherol in RPE cells rescued BCD phenotypes, whereas glucosylceramide reduction did not affect the BCD phenotype. Our data provide evidence that reducing intracellular free cholesterol may have therapeutic efficacy in patients with BCD.
Collapse
|
29
|
Ebner L, Gläser A, Bräuer A, Witt M, Wree A, Rolfs A, Frank M, Vollmar B, Kuhla A. Evaluation of Two Liver Treatment Strategies in a Mouse Model of Niemann-Pick-Disease Type C1. Int J Mol Sci 2018; 19:ijms19040972. [PMID: 29587349 PMCID: PMC5979582 DOI: 10.3390/ijms19040972] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2018] [Revised: 03/14/2018] [Accepted: 03/20/2018] [Indexed: 11/16/2022] Open
Abstract
Niemann–Pick-disease type C1 (NPC1) is an autosomal-recessive cholesterol-storage disorder. Besides other symptoms, NPC1 patients develop liver dysfunction and hepatosplenomegaly. The mechanisms of hepatomegaly and alterations of lipid metabolism-related genes in NPC1 disease are still poorly understood. Here, we used an NPC1 mouse model to study an additive hepatoprotective effect of a combination of 2-hydroxypropyl-β-cyclodextrin (HPβCD), miglustat and allopregnanolone (combination therapy) with the previously established monotherapy using HPβCD. We examined transgene effects as well as treatment effects on liver morphology and hepatic lipid metabolism, focusing on hepatic cholesterol transporter genes. Livers of Npc1−/− mice showed hepatic cholesterol sequestration with consecutive liver injury, an increase of lipogenetic gene expression, e.g., HMG-CoA, a decrease of lipolytic gene expression, e.g., pparα and acox1, and a decrease of lipid transporter gene expression, e.g., acat1, abca1 and fatp2. Both, combination therapy and monotherapy, led to a reduction of hepatic lipids and an amelioration of NPC1 liver disease symptoms. Monotherapy effects were related to pparα- and acox1-associated lipolysis/β-oxidation and to fatp2-induced fatty acid transport, whereas the combination therapy additionally increased the cholesterol transport via abca1 and apoE. However, HPβCD monotherapy additionally increased cholesterol synthesis as indicated by a marked increase of the HMG-CoA and srebp-2 mRNA expression, probably as a result of increased hepatocellular proliferation.
Collapse
Affiliation(s)
- Lynn Ebner
- Institute for Experimental Surgery, Rostock University Medical Center, Schillingallee 69a, 18057 Rostock, Germany.
| | - Anne Gläser
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstraße 9, 18057 Rostock, Germany.
| | - Anja Bräuer
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstraße 9, 18057 Rostock, Germany.
| | - Martin Witt
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstraße 9, 18057 Rostock, Germany.
| | - Andreas Wree
- Institute of Anatomy, Rostock University Medical Center, Gertrudenstraße 9, 18057 Rostock, Germany.
| | - Arndt Rolfs
- Albrecht Kossel Institute for Neuroregeneration, Rostock University Medical Center, Gehlsheimer Straße 20, 18147 Rostock, Germany.
| | - Marcus Frank
- Medical Biology and Electron Microscopy Center, Rostock University Medical Center, 18057 Rostock, Germany.
| | - Brigitte Vollmar
- Institute for Experimental Surgery, Rostock University Medical Center, Schillingallee 69a, 18057 Rostock, Germany.
| | - Angela Kuhla
- Institute for Experimental Surgery, Rostock University Medical Center, Schillingallee 69a, 18057 Rostock, Germany.
| |
Collapse
|
30
|
Crumling MA, King KA, Duncan RK. Cyclodextrins and Iatrogenic Hearing Loss: New Drugs with Significant Risk. Front Cell Neurosci 2017; 11:355. [PMID: 29163061 PMCID: PMC5676048 DOI: 10.3389/fncel.2017.00355] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2017] [Accepted: 10/26/2017] [Indexed: 12/18/2022] Open
Abstract
Cyclodextrins are a family of cyclic oligosaccharides with widespread usage in medicine, industry and basic sciences owing to their ability to solubilize and stabilize guest compounds. In medicine, cyclodextrins primarily act as a complexing vehicle and consequently serve as powerful drug delivery agents. Recently, uncomplexed cyclodextrins have emerged as potent therapeutic compounds in their own right, based on their ability to sequester and mobilize cellular lipids. In particular, 2-hydroxypropyl-β-cyclodextrin (HPβCD) has garnered attention because of its cholesterol chelating properties, which appear to treat a rare neurodegenerative disorder and to promote atherosclerosis regression related to stroke and heart disease. Despite the potential health benefits, use of HPβCD has been linked to significant hearing loss in several species, including humans. Evidence in mice supports a rapid onset of hearing loss that is dose-dependent. Ototoxicity can occur following central or peripheral drug delivery, with either route resulting in the preferential loss of cochlear outer hair cells (OHCs) within hours of dosing. Inner hair cells and spiral ganglion cells are spared at doses that cause ~85% OHC loss; additionally, no other major organ systems appear adversely affected. Evidence from a first-to-human phase 1 clinical trial mirrors animal studies to a large extent, indicating rapid onset and involvement of OHCs. All patients in the trial experienced some permanent hearing loss, although a temporary loss of function can be observed acutely following drug delivery. The long-term impact of HPβCD use as a maintenance drug, and the mechanism(s) of ototoxicity, are unknown. β-cyclodextrins preferentially target membrane cholesterol, but other lipid species and proteins may be directly or indirectly involved. Moreover, as cholesterol is ubiquitous in cell membranes, it remains unclear why OHCs are preferentially susceptible to HPβCD. It is possible that HPβCD acts upon several targets—for example, ion channels, tight junctions (TJ), membrane integrity, and bioenergetics—that collectively increase the sensitivity of OHCs over other cell types.
Collapse
Affiliation(s)
- Mark A Crumling
- Department of Otolaryngology-Head & Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI, United States
| | - Kelly A King
- Audiology Unit, Otolaryngology Branch, National Institute on Deafness and Other Communication Disorders, National Institutes of Health, Bethesda, MD, United States
| | - R Keith Duncan
- Department of Otolaryngology-Head & Neck Surgery, Kresge Hearing Research Institute, University of Michigan, Ann Arbor, MI, United States
| |
Collapse
|
31
|
Bakke SS, Aune MH, Niyonzima N, Pilely K, Ryan L, Skjelland M, Garred P, Aukrust P, Halvorsen B, Latz E, Damås JK, Mollnes TE, Espevik T. Cyclodextrin Reduces Cholesterol Crystal–Induced Inflammation by Modulating Complement Activation. THE JOURNAL OF IMMUNOLOGY 2017; 199:2910-2920. [DOI: 10.4049/jimmunol.1700302] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Accepted: 08/09/2017] [Indexed: 11/19/2022]
|
32
|
Reversal of Pathologic Lipid Accumulation in NPC1-Deficient Neurons by Drug-Promoted Release of LAMP1-Coated Lamellar Inclusions. J Neurosci 2017; 36:8012-25. [PMID: 27466344 DOI: 10.1523/jneurosci.0900-16.2016] [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: 03/18/2016] [Accepted: 06/15/2016] [Indexed: 02/05/2023] Open
Abstract
UNLABELLED Aging and pathologic conditions cause intracellular aggregation of macromolecules and the dysfunction and degeneration of neurons, but the mechanisms are largely unknown. Prime examples are lysosomal storage disorders such as Niemann-Pick type C (NPC) disease, where defects in the endosomal-lysosomal protein NPC1 or NPC2 cause intracellular accumulation of unesterified cholesterol and other lipids leading to neurodegeneration and fatal neurovisceral symptoms. Here, we investigated the impact of NPC1 deficiency on rodent neurons using pharmacologic and genetic models of the disease. Improved ultrastructural detection of lipids and correlative light and electron microscopy identified lamellar inclusions as the subcellular site of cholesterol accumulation in neurons with impaired NPC1 activity. Immunogold labeling combined with transmission electron microscopy revealed the presence of CD63 on internal lamellae and of LAMP1 on the membrane surrounding the inclusions, indicating their origins from intraluminal vesicles of late endosomes and of a lysosomal compartment, respectively. Lamellar inclusions contained cell-intrinsic cholesterol and surface-labeled GM1, indicating the incorporation of plasma membrane components. Scanning electron microscopy revealed that the therapeutic drug candidate β-cyclodextrin induces the subplasmalemmal location of lamellar inclusions and their subsequent release to the extracellular space. In parallel, β-cyclodextrin mediated the NPC1-independent redistribution of cholesterol within neurons and thereby abolished a deleterious cycle of enhanced cholesterol synthesis and its intracellular accumulation, which was indicated by neuron-specific transcript analysis. Our study provides new mechanistic insight into the pathologic aggregation of macromolecules in neurons and suggests exocytosis as cellular target for its therapeutic reversal. SIGNIFICANCE STATEMENT Many neurodegenerative diseases involve pathologic accumulation of molecules within neurons, but the subcellular location and the cellular impact are often unknown and therapeutic approaches lacking. We investigated these questions in the lysosomal storage disorder Niemann-Pick type C (NPC), where a defect in intracellular cholesterol transport causes loss of neurons and fatal neurovisceral symptoms. Here, we identify lamellar inclusions as the subcellular site of lipid accumulation in neurons, we uncover a vicious cycle of cholesterol synthesis and accretion, which may cause gradual neurodegeneration, and we reveal how β-cyclodextrin, a potential therapeutic drug, reverts these changes. Our study provides new mechanistic insight in NPC disease and uncovers new targets for therapeutic approaches.
Collapse
|
33
|
Meyer A, Wree A, Günther R, Holzmann C, Schmitt O, Rolfs A, Witt M. Increased Regenerative Capacity of the Olfactory Epithelium in Niemann-Pick Disease Type C1. Int J Mol Sci 2017; 18:ijms18040777. [PMID: 28383485 PMCID: PMC5412361 DOI: 10.3390/ijms18040777] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2017] [Revised: 03/19/2017] [Accepted: 03/20/2017] [Indexed: 11/28/2022] Open
Abstract
Niemann–Pick disease type C1 (NPC1) is a fatal neurovisceral lysosomal lipid storage disorder. The mutation of the NPC1 protein affects the homeostasis and transport of cholesterol and glycosphingolipids from late endosomes/lysosomes to the endoplasmic reticulum resulting in progressive neurodegeneration. Since olfactory impairment is one of the earliest symptoms in many neurodegenerative disorders, we focused on alterations of the olfactory epithelium in an NPC1 mouse model. Previous findings revealed severe morphological and immunohistochemical alterations in the olfactory system of NPC1−/− mutant mice compared with healthy controls (NPC1+/+). Based on immunohistochemical evaluation of the olfactory epithelium, we analyzed the impact of neurodegeneration in the olfactory epithelium of NPC1−/− mice and observed considerable loss of mature olfactory receptor neurons as well as an increased number of proliferating and apoptotic cells. Additionally, after administration of two different therapy approaches using either a combination of miglustat, 2-hydroxypropyl-β-cyclodextrin (HPβCD) and allopregnanolone or a monotherapy with HPβCD, we recorded a remarkable reduction of morphological damages in NPC1−/− mice and an up to four-fold increase of proliferating cells within the olfactory epithelium. Numbers of mature olfactory receptor neurons doubled after both therapy approaches. Interestingly, we also observed therapy-induced alterations in treated NPC1+/+ controls. Thus, olfactory testing may provide useful information to monitor pharmacologic treatment approaches in human NPC1.
Collapse
Affiliation(s)
- Anja Meyer
- Institute of Anatomy, University of Rostock, 18057 Rostock, Germany.
| | - Andreas Wree
- Institute of Anatomy, University of Rostock, 18057 Rostock, Germany.
| | - René Günther
- Institute of Anatomy, University of Rostock, 18057 Rostock, Germany.
| | - Carsten Holzmann
- Institute of Medical Genetics, Rostock University Medical Center, 18057 Rostock, Germany.
| | - Oliver Schmitt
- Institute of Anatomy, University of Rostock, 18057 Rostock, Germany.
| | - Arndt Rolfs
- Albrecht-Kossel Institute for Neuroregeneration, Rostock University Medical Center, 18147 Rostock, Germany.
| | - Martin Witt
- Institute of Anatomy, University of Rostock, 18057 Rostock, Germany.
| |
Collapse
|
34
|
Mendelsohn AR, Larrick JW. Preclinical Reversal of Atherosclerosis by FDA-Approved Compound that Transforms Cholesterol into an Anti-Inflammatory "Prodrug". Rejuvenation Res 2017; 19:252-5. [PMID: 27241174 DOI: 10.1089/rej.2016.1849] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Although atherosclerosis is treatable with lipid-lowering drugs, not all patients respond. Hydroxypropyl-beta-cyclodextrin (CD) is an FDA-approved compound for solubilizing, capturing, and delivering lipophilic drugs in humans. Zimmer et al. report that CD mediates regression of atherosclerotic plaques in two mouse models by solubilizing cholesterol crystals (CCs), and promoting metabolism of CCs into water-soluble 27-hydroxycholesterol, which, in turn, activates anti-inflammatory LXR receptor target genes, promotes active and passive efflux of cholesterol from macrophages, and increases metabolic processing of cholesterol. In effect, CD inverts the role of its cargo, cholesterol, from inflammatory to anti-inflammatory by converting cholesterol into a "prodrug" that when modified to 27-hydroxycholesterol reduces atherosclerosis. This mechanism defines a new class of pharmaceuticals, "inverters": compounds that cause innate biomolecules to act opposite to their normal function. However, chronic CD treatment in animal models damages auditory cells, which must be addressed before CD can be developed as a systemic drug for atherosclerosis.
Collapse
Affiliation(s)
- Andrew R Mendelsohn
- 1 Panorama Research Institute , Sunnyvale, California.,2 Regenerative Sciences Institute , Sunnyvale, California
| | - James W Larrick
- 1 Panorama Research Institute , Sunnyvale, California.,2 Regenerative Sciences Institute , Sunnyvale, California
| |
Collapse
|
35
|
Abstract
Beta-cyclodextrin (β-CD) has been applied as drug/food carriers or potential drugs for treating some diseases. Most recently, some evidence indicated that methyl-β-cyclodextrin (MβCD) and 2-hydroxypropyl-β-cyclodextrin (2-HPβCD), two major derivatives of β-CD, may inhibit atherogenesis, implying that cyclodextrins also can be potential drugs for treating atherosclerosis. It is well known that modification (e.g. oxidation) of low-density lipoprotein (LDL) is one of the most critical steps of atherogenesis. Lipoxygenase, an enzyme able to be expressed by atherosclerosis-related vascular cells, is generally regarded as a possible in vivo agent of LDL oxidation. In this study, the effects of MβCD on LDL oxidation induced by lipoxygenase were investigated by measuring the electrophoretic mobility, conjugated diene formation, malondialdehyde (MDA) production, and amino group blockage of LDL. We found that the lipids depleted from LDL by MβCD could be oxygenated more readily by lipoxygenase whereas the lipoxygenase-induced oxidation of the remaining lipid-depleted LDL decreased. The data imply that MβCD has an inhibitory effect on lipoxygenase-induced LDL oxidation and probably helps to inhibit atherogenesis.
Collapse
Affiliation(s)
- Meiying Ao
- College of Life Sciences, Nanchang University
| | | |
Collapse
|
36
|
Newton J, Hait NC, Maceyka M, Colaco A, Maczis M, Wassif CA, Cougnoux A, Porter FD, Milstien S, Platt N, Platt FM, Spiegel S. FTY720/fingolimod increases NPC1 and NPC2 expression and reduces cholesterol and sphingolipid accumulation in Niemann-Pick type C mutant fibroblasts. FASEB J 2017; 31:1719-1730. [PMID: 28082351 DOI: 10.1096/fj.201601041r] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2016] [Accepted: 01/03/2017] [Indexed: 11/11/2022]
Abstract
Niemann-Pick type C (NPC) disease is a fatal neurodegenerative disorder caused by mutations in NPC1 or NPC2 with decreased functions leading to lysosomal accumulation of cholesterol and sphingolipids. FTY720/fingolimod, used for treatment of multiple sclerosis, is phosphorylated by nuclear sphingosine kinase 2, and its active phosphorylated form (FTY720-P) is an inhibitor of class I histone deacetylases. In this study, administration of clinically relevant doses of FTY720 to mice increased expression of NPC1 and -2 in brain and liver and decreased cholesterol in an SphK2-dependent manner. FTY720 greatly increased expression of NPC1 and -2 in human NPC1 mutant fibroblasts that correlated with formation of FTY720-P and significantly reduced the accumulation of cholesterol and glycosphingolipids. In agreement with this finding, FTY720 pretreatment of human NPC1 mutant fibroblasts restored transport of the cholera toxin B subunit, which binds ganglioside GM1, to the Golgi apparatus. Together, these findings suggest that FTY720 administration can ameliorate cholesterol and sphingolipid storage and trafficking defects in NPC1 mutant fibroblasts. Because neurodegeneration is the main clinical feature of NPC disease, and FTY720 accumulates in the CNS and has several advantages over available histone deacetylase inhibitors now in clinical trials, our work provides a potential opportunity for treatment of this incurable disease.-Newton, J., Hait, N. C., Maceyka, M., Colaco, A., Maczis, M., Wassif, C. A., Cougnoux, A., Porter, F. D., Milstien, S., Platt, N., Platt, F. M., Spiegel, S. FTY720/fingolimod increases NPC1 and NPC2 expression and reduces cholesterol and sphingolipid accumulation in Niemann-Pick type C mutant fibroblasts.
Collapse
Affiliation(s)
- Jason Newton
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Nitai C Hait
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Michael Maceyka
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Alexandria Colaco
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom; and
| | - Melissa Maczis
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Christopher A Wassif
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Antony Cougnoux
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Forbes D Porter
- Division of Translational Medicine, Eunice Kennedy Shriver National Institute of Child Health and Human Development, National Institutes of Health, Bethesda, Maryland, USA
| | - Sheldon Milstien
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA
| | - Nicholas Platt
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom; and
| | - Frances M Platt
- Department of Pharmacology, University of Oxford, Oxford, United Kingdom; and
| | - Sarah Spiegel
- Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, Richmond, Virginia, USA;
| |
Collapse
|
37
|
Munkacsi AB, Hammond N, Schneider RT, Senanayake DS, Higaki K, Lagutin K, Bloor SJ, Ory DS, Maue RA, Chen FW, Hernandez-Ono A, Dahlson N, Repa JJ, Ginsberg HN, Ioannou YA, Sturley SL. Normalization of Hepatic Homeostasis in the Npc1nmf164 Mouse Model of Niemann-Pick Type C Disease Treated with the Histone Deacetylase Inhibitor Vorinostat. J Biol Chem 2016; 292:4395-4410. [PMID: 28031458 DOI: 10.1074/jbc.m116.770578] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2016] [Revised: 12/21/2016] [Indexed: 12/31/2022] Open
Abstract
Niemann-Pick type C (NP-C) disease is a fatal genetic lipidosis for which there is no Food and Drug Administration (FDA)-approved therapy. Vorinostat, an FDA-approved inhibitor of histone deacetylases, ameliorates lysosomal lipid accumulation in cultured NP-C patient fibroblasts. To assess the therapeutic potential of histone deacetylase inhibition, we pursued these in vitro observations in two murine models of NP-C disease. Npc1nmf164 mice, which express a missense mutation in the Npc1 gene, were treated intraperitoneally, from weaning, with the maximum tolerated dose of vorinostat (150 mg/kg, 5 days/week). Disease progression was measured via gene expression, liver function and pathology, serum and tissue lipid levels, body weight, and life span. Transcriptome analyses of treated livers indicated multiple changes consistent with reversal of liver dysfunction that typifies NP-C disease. Significant improvements in liver pathology and function were achieved by this treatment regimen; however, NPC1 protein maturation and levels, disease progression, weight loss, and animal morbidity were not detectably altered. Vorinostat concentrations were >200 μm in the plasma compartment of treated animals but were almost 100-fold lower in brain tissue. Apolipoprotein B metabolism and the expression of key components of lipid homeostasis in primary hepatocytes from null (Npc1-/-) and missense (Npc1nmf164 ) mutant mice were altered by vorinostat treatment, consistent with a response by these cells independent of the status of the Npc1 locus. These results suggest that HDAC inhibitors have utility to treat visceral NP-C disease. However, it is clear that improved blood-brain barrier penetration will be required to alleviate the neurological symptoms of human NP-C disease.
Collapse
Affiliation(s)
- Andrew B Munkacsi
- From the School of Biological Sciences and .,Centre for Biodiscovery, Victoria University of Wellington, Wellington 6012, New Zealand
| | | | | | | | - Katsumi Higaki
- the Division of Functional Genomics, Research Center for Bioscience and Technology, Tottori University, Yonago 683-8503, Japan
| | | | | | - Daniel S Ory
- the Department of Medicine, Washington University School of Medicine, St. Louis, Missouri 63110
| | - Robert A Maue
- the Department of Physiology and Neurobiology and the Department of Biochemistry, Geisel School of Medicine at Dartmouth, Hanover, New Hampshire 03755
| | - Fannie W Chen
- the Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York 10029
| | | | - Nicole Dahlson
- the Departments of Physiology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, and
| | - Joyce J Repa
- the Departments of Physiology and Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas 75390, and
| | | | - Yiannis A Ioannou
- the Department of Genetics and Genomic Sciences, Mount Sinai School of Medicine, New York, New York 10029
| | - Stephen L Sturley
- the Department of Genetics and Development, Columbia University Medical Center, New York, New York 10032
| |
Collapse
|
38
|
Zimmer S, Grebe A, Bakke SS, Bode N, Halvorsen B, Ulas T, Skjelland M, De Nardo D, Labzin LI, Kerksiek A, Hempel C, Heneka MT, Hawxhurst V, Fitzgerald ML, Trebicka J, Björkhem I, Gustafsson JÅ, Westerterp M, Tall AR, Wright SD, Espevik T, Schultze JL, Nickenig G, Lütjohann D, Latz E. Cyclodextrin promotes atherosclerosis regression via macrophage reprogramming. Sci Transl Med 2016; 8:333ra50. [PMID: 27053774 DOI: 10.1126/scitranslmed.aad6100] [Citation(s) in RCA: 265] [Impact Index Per Article: 29.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2015] [Accepted: 02/18/2016] [Indexed: 12/12/2022]
Abstract
Atherosclerosis is an inflammatory disease linked to elevated blood cholesterol concentrations. Despite ongoing advances in the prevention and treatment of atherosclerosis, cardiovascular disease remains the leading cause of death worldwide. Continuous retention of apolipoprotein B-containing lipoproteins in the subendothelial space causes a local overabundance of free cholesterol. Because cholesterol accumulation and deposition of cholesterol crystals (CCs) trigger a complex inflammatory response, we tested the efficacy of the cyclic oligosaccharide 2-hydroxypropyl-β-cyclodextrin (CD), a compound that increases cholesterol solubility in preventing and reversing atherosclerosis. We showed that CD treatment of murine atherosclerosis reduced atherosclerotic plaque size and CC load and promoted plaque regression even with a continued cholesterol-rich diet. Mechanistically, CD increased oxysterol production in both macrophages and human atherosclerotic plaques and promoted liver X receptor (LXR)-mediated transcriptional reprogramming to improve cholesterol efflux and exert anti-inflammatory effects. In vivo, this CD-mediated LXR agonism was required for the antiatherosclerotic and anti-inflammatory effects of CD as well as for augmented reverse cholesterol transport. Because CD treatment in humans is safe and CD beneficially affects key mechanisms of atherogenesis, it may therefore be used clinically to prevent or treat human atherosclerosis.
Collapse
Affiliation(s)
- Sebastian Zimmer
- Medizinische Klinik und Poliklinik II, University Hospital Bonn, 53105 Bonn, Germany
| | - Alena Grebe
- Institute of Innate Immunity, University Hospital Bonn, 53127 Bonn, Germany
| | - Siril S Bakke
- Institute of Innate Immunity, University Hospital Bonn, 53127 Bonn, Germany. German Center of Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany. Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, 7489 Trondheim, Norway
| | - Niklas Bode
- Medizinische Klinik und Poliklinik II, University Hospital Bonn, 53105 Bonn, Germany
| | - Bente Halvorsen
- Research Institute of Internal Medicine, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway
| | - Thomas Ulas
- Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Mona Skjelland
- Department of Neurology, Oslo University Hospital Rikshospitalet, 0424 Oslo, Norway
| | - Dominic De Nardo
- Institute of Innate Immunity, University Hospital Bonn, 53127 Bonn, Germany. Inflammation Division, The Walter and Eliza Hall Institute of Medical Research, Parkville, Victoria 3052, Australia. Department of Medical Biology, The University of Melbourne, Parkville, Victoria 3010, Australia
| | - Larisa I Labzin
- Institute of Innate Immunity, University Hospital Bonn, 53127 Bonn, Germany
| | - Anja Kerksiek
- Institute of Clinical Chemistry und Clinical Pharmacology, University Hospital Bonn, 53105 Bonn, Germany
| | | | - Michael T Heneka
- Clinic and Polyclinic for Neurology, University Hospital Bonn, 53105 Bonn, Germany
| | - Victoria Hawxhurst
- Lipid Metabolism Unit, Center for Computational and Integrative Biology, Boston, MA 02114, USA
| | - Michael L Fitzgerald
- Lipid Metabolism Unit, Center for Computational and Integrative Biology, Boston, MA 02114, USA
| | - Jonel Trebicka
- Medizinische Klinik und Poliklinik I, University Hospital Bonn, 53105 Bonn, Germany. Faculty of Health Sciences, University of Southern Denmark Campusvej 55, DK-5230 Odense M, Denmark
| | - Ingemar Björkhem
- Division of Clinical Chemistry, Karolinska Institutet, Huddinge University Hospital, 141 86 Huddinge, Sweden
| | - Jan-Åke Gustafsson
- Center for Nuclear Receptors and Cell Signaling, University of Houston, Houston, TX 77004, USA
| | - Marit Westerterp
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | - Alan R Tall
- Department of Medicine, Columbia University, New York, NY 10032, USA
| | | | - Terje Espevik
- Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, 7489 Trondheim, Norway
| | - Joachim L Schultze
- German Center of Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany. Genomics and Immunoregulation, Life and Medical Sciences Institute, University of Bonn, 53115 Bonn, Germany
| | - Georg Nickenig
- Medizinische Klinik und Poliklinik II, University Hospital Bonn, 53105 Bonn, Germany
| | - Dieter Lütjohann
- Institute of Clinical Chemistry und Clinical Pharmacology, University Hospital Bonn, 53105 Bonn, Germany
| | - Eicke Latz
- Institute of Innate Immunity, University Hospital Bonn, 53127 Bonn, Germany. German Center of Neurodegenerative Diseases (DZNE), 53127 Bonn, Germany. Centre of Molecular Inflammation Research, Norwegian University of Science and Technology, 7489 Trondheim, Norway. Department of Infectious Diseases and Immunology, University of Massachusetts Medical School, Worcester, MA 01605, USA.
| |
Collapse
|
39
|
Long Y, Xu M, Li R, Dai S, Beers J, Chen G, Soheilian F, Baxa U, Wang M, Marugan JJ, Muro S, Li Z, Brady R, Zheng W. Induced Pluripotent Stem Cells for Disease Modeling and Evaluation of Therapeutics for Niemann-Pick Disease Type A. Stem Cells Transl Med 2016; 5:1644-1655. [PMID: 27484861 PMCID: PMC5189647 DOI: 10.5966/sctm.2015-0373] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2015] [Accepted: 05/13/2016] [Indexed: 11/16/2022] Open
Abstract
Niemann-Pick disease type A (NPA)-induced pluripotent stem cells from patient dermal fibroblasts were differentiated into neural stem cells. By using the differentiated NPA neuronal cells as a cell-based disease model system, α-tocopherol, δ-tocopherol, and hydroxypropyl-β-cyclodextrin significantly reduced sphingomyelin accumulation in these NPA neuronal cells. This cell-based NPA model can be used for further study of disease pathophysiology and for high-throughput screening of compound libraries to identify lead compounds for drug development. Niemann-Pick disease type A (NPA) is a lysosomal storage disease caused by mutations in the SMPD1 gene that encodes acid sphingomyelinase (ASM). Deficiency in ASM function results in lysosomal accumulation of sphingomyelin and neurodegeneration. Currently, there is no effective treatment for NPA. To accelerate drug discovery for treatment of NPA, we generated induced pluripotent stem cells from two patient dermal fibroblast lines and differentiated them into neural stem cells. The NPA neural stem cells exhibit a disease phenotype of lysosomal sphingomyelin accumulation and enlarged lysosomes. By using this disease model, we also evaluated three compounds that reportedly reduced lysosomal lipid accumulation in Niemann-Pick disease type C as well as enzyme replacement therapy with ASM. We found that α-tocopherol, δ-tocopherol, hydroxypropyl-β-cyclodextrin, and ASM reduced sphingomyelin accumulation and enlarged lysosomes in NPA neural stem cells. Therefore, the NPA neural stem cells possess the characteristic NPA disease phenotype that can be ameliorated by tocopherols, cyclodextrin, and ASM. Our results demonstrate the efficacies of cyclodextrin and tocopherols in the NPA cell-based model. Our data also indicate that the NPA neural stem cells can be used as a new cell-based disease model for further study of disease pathophysiology and for high-throughput screening to identify new lead compounds for drug development. Significance Currently, there is no effective treatment for Niemann-Pick disease type A (NPA). To accelerate drug discovery for treatment of NPA, NPA-induced pluripotent stem cells were generated from patient dermal fibroblasts and differentiated into neural stem cells. By using the differentiated NPA neuronal cells as a cell-based disease model system, α-tocopherol, δ-tocopherol, and hydroxypropyl-β-cyclodextrin significantly reduced sphingomyelin accumulation in these NPA neuronal cells. Therefore, this cell-based NPA model can be used for further study of disease pathophysiology and for high-throughput screening of compound libraries to identify lead compounds for drug development.
Collapse
Affiliation(s)
- Yan Long
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Miao Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Rong Li
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Sheng Dai
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
- Sir Run Run Shaw Hospital, Zhejiang University School of Medicine, Hangzhou, People's Republic of China
| | - Jeanette Beers
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
| | - Guokai Chen
- Center for Molecular Medicine, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, Maryland, USA
- Faculty of Health Sciences, University of Macau, Macau, People's Republic of China
| | - Ferri Soheilian
- Electron Microscopy Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Ulrich Baxa
- Electron Microscopy Laboratory, Cancer Research Technology Program, Leidos Biomedical Research, Inc., Frederick National Laboratory for Cancer Research, Frederick, Maryland, USA
| | - Mengqiao Wang
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Juan J Marugan
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| | - Silvia Muro
- Institute for Bioscience and Biotechnology Research, University of Maryland, College Park, Maryland, USA
- Fischell Department of Bioengineering, University of Maryland, College Park, Maryland, USA
| | - Zhiyuan Li
- Guangzhou Institutes of Biomedicine and Health, Chinese Academy of Sciences, Guangzhou, People's Republic of China
| | - Roscoe Brady
- National Institute of Neurological Disorders and Stroke, National Institutes of Health, Bethesda, Maryland, USA
| | - Wei Zheng
- National Center for Advancing Translational Sciences, National Institutes of Health, Bethesda, Maryland, USA
| |
Collapse
|
40
|
Bradbury A, Bagel J, Sampson M, Farhat N, Ding W, Swain G, Prociuk M, O'Donnell P, Drobatz K, Gurda B, Wassif C, Remaley A, Porter F, Vite C. Cerebrospinal Fluid Calbindin D Concentration as a Biomarker of Cerebellar Disease Progression in Niemann-Pick Type C1 Disease. J Pharmacol Exp Ther 2016; 358:254-61. [PMID: 27307499 PMCID: PMC4959104 DOI: 10.1124/jpet.116.232975] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2016] [Accepted: 06/06/2016] [Indexed: 01/29/2023] Open
Abstract
Niemann-Pick type C (NPC) 1 disease is a rare, inherited, neurodegenerative disease. Clear evidence of the therapeutic efficacy of 2-hydroxypropyl-β-cyclodextrin (HPβCD) in animal models resulted in the initiation of a phase I/IIa clinical trial in 2013 and a phase IIb/III trial in 2015. With clinical trials ongoing, validation of a biomarker to track disease progression and serve as a supporting outcome measure of therapeutic efficacy has become compulsory. In this study, we evaluated calcium-binding protein calbindin D-28K (calbindin) concentrations in the cerebrospinal fluid (CSF) as a biomarker of NPC1 disease. In the naturally occurring feline model, CSF calbindin was significantly elevated at 3 weeks of age, prior to the onset of cerebellar dysfunction, and steadily increased to >10-fold over normal at end-stage disease. Biweekly intrathecal administration of HPβCD initiated prior to the onset of neurologic dysfunction completely normalized CSF calbindin in NPC1 cats at all time points analyzed when followed up to 78 weeks of age. Initiation of HPβCD after the onset of clinical signs (16 weeks of age) resulted in a delayed reduction of calbindin levels in the CSF. Evaluation of CSF from patients with NPC1 revealed that calbindin concentrations were significantly elevated compared with CSF samples collected from unaffected patients. Off-label treatment of patients with NPC1 with miglustat, an inhibitor of glycosphingolipid biosynthesis, significantly decreased CSF calbindin compared with pretreatment concentrations. These data suggest that the CSF calbindin concentration is a sensitive biomarker of NPC1 disease that could be instrumental as an outcome measure of therapeutic efficacy in ongoing clinical trials.
Collapse
Affiliation(s)
- Allison Bradbury
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Jessica Bagel
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Maureen Sampson
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Nicole Farhat
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Wenge Ding
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Gary Swain
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Maria Prociuk
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Patricia O'Donnell
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Kenneth Drobatz
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Brittney Gurda
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Christopher Wassif
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Alan Remaley
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Forbes Porter
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| | - Charles Vite
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, Pennsylvania (A.B., J.B., W.D., G.S., M.P., P.O., K.D., B.G., C.V.); Division of Intramural Research, National Institutes of Health National Heart, Lung, and Blood Institute, Bethesda, Maryland (M.S., A.R.); and Division of Translational Research, National Institutes of Health Eunice Kennedy Shriver National Institute of Child Health and Human Development, Bethesda, Maryland (N.F., C.W., F.P.)
| |
Collapse
|
41
|
Davidson CD, Fishman YI, Puskás I, Szemán J, Sohajda T, McCauliff LA, Sikora J, Storch J, Vanier MT, Szente L, Walkley SU, Dobrenis K. Efficacy and ototoxicity of different cyclodextrins in Niemann-Pick C disease. Ann Clin Transl Neurol 2016; 3:366-80. [PMID: 27231706 PMCID: PMC4863749 DOI: 10.1002/acn3.306] [Citation(s) in RCA: 67] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2015] [Revised: 03/14/2016] [Accepted: 03/16/2016] [Indexed: 01/04/2023] Open
Abstract
OBJECTIVE Niemann-Pick type C (NPC) disease is a fatal, neurodegenerative, lysosomal storage disorder characterized by intracellular accumulation of unesterified cholesterol (UC) and other lipids. While its mechanism of action remains unresolved, administration of 2-hydroxypropyl-β-cyclodextrin (HPβCD) has provided the greatest disease amelioration in animal models but is ototoxic. We evaluated other cyclodextrins (CDs) for treatment outcome and chemical interaction with disease-relevant substrates that could pertain to mechanism. METHODS NPC disease mice treated for 2 weeks with nine different CDs were evaluated for UC, and GM2 and GM3 ganglioside accumulation using immunohisto/cytochemical and biochemical assays. Auditory brainstem responses were determined in wild-type mice administered CDs. CD complexation with UC, gangliosides, and other lipids was quantified. RESULTS Four HPβCDs varying in degrees of substitution, including one currently in clinical trial, showed equivalent storage reduction, while other CDs showed significant differences in relative ototoxicity and efficacy, with reductions similar for the brain and liver. Importantly, HPγCD and two sulfobutylether-CDs showed efficacy with reduced ototoxicity. Complexation studies showed: incomplete correlation between CD efficacy and UC solubilization; an inverse correlation for ganglioside complexation; substantial interaction with several relevant lipids; and association between undesirable increases of UC storage in Kupffer cells and UC solubilization. INTERPRETATION CDs other than HPβCD identified here may provide disease amelioration without ototoxicity and merit long-term treatment studies. While direct interactions of CD-UC are thought central to the mechanism of correction, the data show that this does not strictly correlate with complexation ability and suggest interactions with other NPC disease-relevant substrates should be considered.
Collapse
Affiliation(s)
- Cristin D. Davidson
- Dominick P. Purpura Department of NeuroscienceRose F. Kennedy Center Intellectual and Developmental Disabilities Research CenterAlbert Einstein College of MedicineBronxNew York10461
| | - Yonatan I. Fishman
- Dominick P. Purpura Department of NeuroscienceRose F. Kennedy Center Intellectual and Developmental Disabilities Research CenterAlbert Einstein College of MedicineBronxNew York10461
| | - István Puskás
- CycloLab Cyclodextrin Research & Development Laboratory Ltd.BudapestH‐1097Hungary
| | - Julianna Szemán
- CycloLab Cyclodextrin Research & Development Laboratory Ltd.BudapestH‐1097Hungary
| | - Tamás Sohajda
- CycloLab Cyclodextrin Research & Development Laboratory Ltd.BudapestH‐1097Hungary
| | - Leslie A. McCauliff
- Department of Nutritional Sciences and Rutgers Center for Lipid ResearchRutgers UniversityNew BrunswickNew Jersey08901
| | - Jakub Sikora
- Dominick P. Purpura Department of NeuroscienceRose F. Kennedy Center Intellectual and Developmental Disabilities Research CenterAlbert Einstein College of MedicineBronxNew York10461
- Institute of Inherited Metabolic DisordersFirst Faculty of MedicineCharles University in Prague and General University Hospital in PraguePragueCzech Republic
| | - Judith Storch
- Department of Nutritional Sciences and Rutgers Center for Lipid ResearchRutgers UniversityNew BrunswickNew Jersey08901
| | - Marie T. Vanier
- Institut National de la Santé et de la Recherche MédicaleUnit 820; EA4611 Lyon‐1 UniversityLyonFrance
| | - Lajos Szente
- CycloLab Cyclodextrin Research & Development Laboratory Ltd.BudapestH‐1097Hungary
| | - Steven U. Walkley
- Dominick P. Purpura Department of NeuroscienceRose F. Kennedy Center Intellectual and Developmental Disabilities Research CenterAlbert Einstein College of MedicineBronxNew York10461
| | - Kostantin Dobrenis
- Dominick P. Purpura Department of NeuroscienceRose F. Kennedy Center Intellectual and Developmental Disabilities Research CenterAlbert Einstein College of MedicineBronxNew York10461
| |
Collapse
|
42
|
Ao M, Gan C, Shao W, Zhou X, Chen Y. Effects of cyclodextrins on the structure of LDL and its susceptibility to copper-induced oxidation. Eur J Pharm Sci 2016; 91:183-9. [PMID: 27140842 DOI: 10.1016/j.ejps.2016.04.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/08/2015] [Revised: 04/28/2016] [Accepted: 04/28/2016] [Indexed: 12/19/2022]
Abstract
Cyclodextrins (CDs) have long been widely used as drug/food carriers and were recently developed as drugs for the treatment of diseases (e.g. Niemann-Pick C1 and cancers). It is unknown whether cyclodextrins may influence the structure of low-density lipoprotein (LDL), its susceptibility to oxidation, and atherogenesis. In this study, four widely used cyclodextrins including α-CD, γ-CD, and two derivatives of β-CD (HPβCD and MβCD) were recruited. Interestingly, agarose gel electrophoresis (staining lipid and protein components of LDL with Sudan Black B and Coomassie brilliant blue, respectively but simultaneously) shows that cyclodextrins at relatively high concentrations caused disappearance of the LDL band and/or appearance of an additional protein-free lipid band, implying that cyclodextrins at relatively high concentrations can induce significant electrophoresis-detectable lipid depletion of LDL. Atomic force microscopy (AFM) detected that MβCD (as a representative of cyclodextrins) induced size decrease of LDL particles in a dose-dependent manner, further confirming the lipid depletion effects of cyclodextrins. Moreover, the data from agarose gel electrophoresis, conjugated diene formation, MDA production, and amino group blockage of copper-oxidized LDL show that cyclodextrins can impair LDL susceptibility to oxidation. It implies that cyclodextrins probably help to inhibit atherogenesis by lowering LDL oxidation.
Collapse
Affiliation(s)
- Meiying Ao
- Nanoscale Science and Technology Laboratory, Institute for Advanced Study, Nanchang University, Nanchang, Jiangxi 330031, PR China; Department of Pharmacy, Science and Technology College, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330025, PR China; College of Life Sciences, Nanchang University, Nanchang, Jiangxi 330031, PR China
| | - Chaoye Gan
- Nanoscale Science and Technology Laboratory, Institute for Advanced Study, Nanchang University, Nanchang, Jiangxi 330031, PR China; College of Life Sciences, Nanchang University, Nanchang, Jiangxi 330031, PR China
| | - Wenxiang Shao
- School of Basic Medical Sciences, Jiangxi University of Traditional Chinese Medicine, Nanchang, Jiangxi 330025, PR China
| | - Xing Zhou
- Nanoscale Science and Technology Laboratory, Institute for Advanced Study, Nanchang University, Nanchang, Jiangxi 330031, PR China; College of Life Sciences, Nanchang University, Nanchang, Jiangxi 330031, PR China
| | - Yong Chen
- Nanoscale Science and Technology Laboratory, Institute for Advanced Study, Nanchang University, Nanchang, Jiangxi 330031, PR China; College of Life Sciences, Nanchang University, Nanchang, Jiangxi 330031, PR China.
| |
Collapse
|
43
|
Tanaka Y, Yamada Y, Ishitsuka Y, Matsuo M, Shiraishi K, Wada K, Uchio Y, Kondo Y, Takeo T, Nakagata N, Higashi T, Motoyama K, Arima H, Mochinaga S, Higaki K, Ohno K, Irie T. Efficacy of 2-Hydroxypropyl-β-cyclodextrin in Niemann-Pick Disease Type C Model Mice and Its Pharmacokinetic Analysis in a Patient with the Disease. Biol Pharm Bull 2016; 38:844-51. [PMID: 26027824 DOI: 10.1248/bpb.b14-00726] [Citation(s) in RCA: 63] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Niemann-Pick type C disease (NPC), an autosomal recessive lysosomal storage disorder, is an inherited disease characterized by the accumulation of intracellular unesterified cholesterol. A solubilizing agent of lipophilic compounds, 2-hydroxypropyl-β-cyclodextrin (HPBCD), is an attractive drug candidate against NPC disease. However, establishment of the optimum dosage of HPBCD remains to be determined. In this study, we evaluated the effective dosage of HPBCD in NPC model (Npc1(-/-)) mice, and determined serum HPBCD concentrations. Subcutaneous injection of 1000-4000 mg/kg HPBCD improved the lifespan of Npc1(-/-) mice. In addition, liver injury and cholesterol sequestration were significantly prevented by 4000 mg/kg HPBCD in Npc1(-/-) mice. Serum HPBCD concentrations, when treated at the effective dosages (1000-4000 mg/kg), were approximately 1200-2500 µg/mL at 0.5 h after subcutaneous injection, and blood HPBCD concentrations were immediately eliminated in Npc1(-/-) mice. Furthermore, we examined serum HPBCD concentrations when treated at 40000 mg (approximately 2500 mg/kg) in a patient with NPC. We observed that the effective concentration in the in vivo study using Npc1(-/-) mice was similar to that in the patient. In the patient, systemic clearance and the volume of distribution of HPBCD were in accordance with the glomerular filtration rate and extracellular fluid volume, respectively. These results could provide useful information for developing the optimal dosage regimen for HPBCD therapy when administered intravenously to NPC patients.
Collapse
Affiliation(s)
- Yuta Tanaka
- Department of Clinical Chemistry and Informatics, Graduate School of Pharmaceutical Sciences, Kumamoto University
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Vite CH, Bagel JH, Swain GP, Prociuk M, Sikora TU, Stein VM, O'Donnell P, Ruane T, Ward S, Crooks A, Li S, Mauldin E, Stellar S, De Meulder M, Kao ML, Ory DS, Davidson C, Vanier MT, Walkley SU. Intracisternal cyclodextrin prevents cerebellar dysfunction and Purkinje cell death in feline Niemann-Pick type C1 disease. Sci Transl Med 2015; 7:276ra26. [PMID: 25717099 DOI: 10.1126/scitranslmed.3010101] [Citation(s) in RCA: 160] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Niemann-Pick type C1 (NPC) disease is a lysosomal storage disease caused by mutations in the NPC1 gene, leading to an increase in unesterified cholesterol and several sphingolipids, and resulting in hepatic disease and progressive neurological disease. We show that subcutaneous administration of the pharmaceutical excipient 2-hydroxypropyl-β-cyclodextrin (HPβCD) to cats with NPC disease ameliorated hepatic disease, but doses sufficient to reduce neurological disease resulted in pulmonary toxicity. However, direct administration of HPβCD into the cisterna magna of presymptomatic cats with NPC disease prevented the onset of cerebellar dysfunction for greater than a year and resulted in a reduction in Purkinje cell loss and near-normal concentrations of cholesterol and sphingolipids. Moreover, administration of intracisternal HPβCD to NPC cats with ongoing cerebellar dysfunction slowed disease progression, increased survival time, and decreased the accumulation of brain gangliosides. An increase in hearing threshold was identified as a potential adverse effect. These studies in a feline animal model have provided critical data on efficacy and safety of drug administration directly into the central nervous system that will be important for advancing HPβCD into clinical trials.
Collapse
Affiliation(s)
- Charles H Vite
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | - Jessica H Bagel
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Gary P Swain
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Maria Prociuk
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Tracey U Sikora
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Veronika M Stein
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Patricia O'Donnell
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Therese Ruane
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Sarah Ward
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Alexandra Crooks
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Su Li
- Department of Clinical Studies, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Elizabeth Mauldin
- Department of Pathobiology, School of Veterinary Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA
| | - Susan Stellar
- Janssen Research & Development, LLC, Janssen Pharmaceutical Companies of Johnson and Johnson, Titusville, NJ 08560, USA
| | - Marc De Meulder
- Janssen Research & Development, a division of Janssen Pharmaceutica NV, Janssen Pharmaceutical Companies of Johnson and Johnson, Beerse, Belgium
| | - Mark L Kao
- Janssen Research & Development, LLC, Janssen Pharmaceutical Companies of Johnson and Johnson, Titusville, NJ 08560, USA
| | - Daniel S Ory
- Diabetic Cardiovascular Disease Center, Washington University School of Medicine, St. Louis, MO 63110, USA
| | - Cristin Davidson
- Dominick P. Purpura Department of Neuroscience, Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Marie T Vanier
- INSERM U820; EA4611, Université Claude Bernard Lyon 1, Lyon, France
| | - Steven U Walkley
- Dominick P. Purpura Department of Neuroscience, Rose F. Kennedy Intellectual and Developmental Disabilities Research Center, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| |
Collapse
|
45
|
Lopez AM, Chuang JC, Posey KS, Ohshiro T, Tomoda H, Rudel LL, Turley SD. PRD125, a potent and selective inhibitor of sterol O-acyltransferase 2 markedly reduces hepatic cholesteryl ester accumulation and improves liver function in lysosomal acid lipase-deficient mice. J Pharmacol Exp Ther 2015; 355:159-67. [PMID: 26283692 PMCID: PMC4613965 DOI: 10.1124/jpet.115.227207] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2015] [Accepted: 08/14/2015] [Indexed: 11/22/2022] Open
Abstract
In most organs, the bulk of cholesterol is unesterified, although nearly all possess a varying capability of esterifying cholesterol through the action of either sterol O-acyltransferase (SOAT) 1 or, in the case of hepatocytes and enterocytes, SOAT2. Esterified cholesterol (EC) carried in plasma lipoproteins is hydrolyzed by lysosomal acid lipase (LAL) when they are cleared from the circulation. Loss-of-function mutations in LIPA, the gene that encodes LAL, result in Wolman disease or cholesteryl ester storage disease (CESD). Hepatomegaly and a massive increase in tissue EC levels are hallmark features of both disorders. While these conditions can be corrected with enzyme replacement therapy, the question arose as to whether pharmacological inhibition of SOAT2 might reduce tissue EC accretion in CESD. When weaned at 21 days, Lal(-/-) mice, of either gender, had a whole liver cholesterol content that was 12- to 13-fold more than that of matching Lal(+/+) littermates (23 versus 1.8 mg, respectively). In Lal(-/-) males given the selective SOAT2 inhibitor PRD125 1,11-O-o-methylbenzylidene-7-O-p-cyanobenzoyl-1,7,11-trideacetylpyripyropene A in their diet (∼10 mg/day per kg body weight) from 21 to 53 days, whole liver cholesterol content was 48.6 versus 153.7 mg in untreated 53-day-old Lal(-/-) mice. This difference reflected a 59% reduction in hepatic EC concentration (mg/g), combined with a 28% fall in liver mass. The treated mice also showed a 63% reduction in plasma alanine aminotransferase activity, in parallel with decisive falls in hepatic mRNA expression levels for multiple proteins that reflect macrophage presence and inflammation. These data implicate SOAT2 as a potential target in CESD management.
Collapse
Affiliation(s)
- Adam M Lopez
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas (A.M.L., J-C.C., K.S.P., S.D.T.); Graduate School of Pharmaceutical Science, Kitasato University, Tokyo, Japan (T.O., H.T.); and Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (T.O., L.L.R.)
| | - Jen-Chieh Chuang
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas (A.M.L., J-C.C., K.S.P., S.D.T.); Graduate School of Pharmaceutical Science, Kitasato University, Tokyo, Japan (T.O., H.T.); and Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (T.O., L.L.R.)
| | - Kenneth S Posey
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas (A.M.L., J-C.C., K.S.P., S.D.T.); Graduate School of Pharmaceutical Science, Kitasato University, Tokyo, Japan (T.O., H.T.); and Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (T.O., L.L.R.)
| | - Taichi Ohshiro
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas (A.M.L., J-C.C., K.S.P., S.D.T.); Graduate School of Pharmaceutical Science, Kitasato University, Tokyo, Japan (T.O., H.T.); and Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (T.O., L.L.R.)
| | - Hiroshi Tomoda
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas (A.M.L., J-C.C., K.S.P., S.D.T.); Graduate School of Pharmaceutical Science, Kitasato University, Tokyo, Japan (T.O., H.T.); and Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (T.O., L.L.R.)
| | - Lawrence L Rudel
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas (A.M.L., J-C.C., K.S.P., S.D.T.); Graduate School of Pharmaceutical Science, Kitasato University, Tokyo, Japan (T.O., H.T.); and Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (T.O., L.L.R.)
| | - Stephen D Turley
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, Texas (A.M.L., J-C.C., K.S.P., S.D.T.); Graduate School of Pharmaceutical Science, Kitasato University, Tokyo, Japan (T.O., H.T.); and Department of Pathology, Wake Forest University School of Medicine, Winston-Salem, North Carolina (T.O., L.L.R.)
| |
Collapse
|
46
|
Weekly Treatment of 2-Hydroxypropyl-β-cyclodextrin Improves Intracellular Cholesterol Levels in LDL Receptor Knockout Mice. Int J Mol Sci 2015; 16:21056-69. [PMID: 26404254 PMCID: PMC4613241 DOI: 10.3390/ijms160921056] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Revised: 08/19/2015] [Accepted: 08/20/2015] [Indexed: 12/13/2022] Open
Abstract
Recently, the importance of lysosomes in the context of the metabolic syndrome has received increased attention. Increased lysosomal cholesterol storage and cholesterol crystallization inside macrophages have been linked to several metabolic diseases, such as atherosclerosis and non-alcoholic fatty liver disease (NAFLD). Two-hydroxypropyl-β-cyclodextrin (HP-B-CD) is able to redirect lysosomal cholesterol to the cytoplasm in Niemann-Pick type C1 disease, a lysosomal storage disorder. We hypothesize that HP-B-CD ameliorates liver cholesterol and intracellular cholesterol levels inside Kupffer cells (KCs). Hyperlipidemic low-density lipoprotein receptor knockout (Ldlr−/−) mice were given weekly, subcutaneous injections with HP-B-CD or control PBS. In contrast to control injections, hyperlipidemic mice treated with HP-B-CD demonstrated a shift in intracellular cholesterol distribution towards cytoplasmic cholesteryl ester (CE) storage and a decrease in cholesterol crystallization inside KCs. Compared to untreated hyperlipidemic mice, the foamy KC appearance and liver cholesterol remained similar upon HP-B-CD administration, while hepatic campesterol and 7α-hydroxycholesterol levels were back increased. Thus, HP-B-CD could be a useful tool to improve intracellular cholesterol levels in the context of the metabolic syndrome, possibly through modulation of phyto- and oxysterols, and should be tested in the future. Additionally, these data underline the existence of a shared etiology between lysosomal storage diseases and NAFLD.
Collapse
|
47
|
Lopez AM, Terpack SJ, Posey KS, Liu B, Ramirez CM, Turley SD. Systemic administration of 2-hydroxypropyl-β-cyclodextrin to symptomatic Npc1-deficient mice slows cholesterol sequestration in the major organs and improves liver function. Clin Exp Pharmacol Physiol 2015; 41:780-7. [PMID: 25115571 DOI: 10.1111/1440-1681.12285] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2014] [Revised: 06/10/2014] [Accepted: 07/04/2014] [Indexed: 11/27/2022]
Abstract
In Niemann-Pick type C (NPC) disease, loss-of-function mutations in either NPC1 or NPC2 result in progressive accumulation of unesterified cholesterol (UC) and glycosphingolipids in all organs, leading to neurodegeneration, pulmonary dysfunction and sometimes liver failure. There is no cure for this disorder. Studies using primarily NPC mouse models have shown that systemic administration of 2-hydroxypropyl-β-cyclodextrin (2HPβCD), starting in early neonatal life, diminishes UC accumulation in most organs, slows disease progression and extends lifespan. The key question now is whether delaying the start of 2HPβCD treatment until early adulthood, when the amount of entrapped UC throughout the body is markedly elevated, has any of the benefits found when treatment begins at 7 days of age. In the present study, Npc1(-/-) and Npc1(+/+) mice were given saline or 2HPβCD subcutaneously at 49, 56, 63 and 70 days of age, with measurements of organ weights, liver function tests and tissue cholesterol levels performed at 77 days. In Npc1(-/-) mice, treatment with 2HPβCD from 49 days reduced whole-liver cholesterol content at 77 days from 33.0 ± 1.0 to 9.1 ± 0.5 mg/organ. Comparable improvements were seen in other organs, such as the spleen, and in the animal as a whole. There was a transient increase in biliary cholesterol concentration in Npc1(-/-) mice after 2HPβCD. Plasma alanine aminotransferase and aspartate aminotransferase activities in 77-day-old 2HPβCD-treated Npc1(-/-) mice were reduced compared with saline-treated controls. The lifespan of Npc1(-/-) mice given 2HPβCD marginally exceeded that of the saline-treated controls (99 ± 1.1 vs 94 ± 1.4 days, respectively; P < 0.05). Thus, 2HPβCD is effective in mobilizing entrapped cholesterol in late-stage NPC disease leading to improved liver function.
Collapse
Affiliation(s)
- Adam M Lopez
- Department of Internal Medicine, University of Texas Southwestern Medical Center, Dallas, TX, USA
| | | | | | | | | | | |
Collapse
|
48
|
Hearing Loss and Otopathology Following Systemic and Intracerebroventricular Delivery of 2-Hydroxypropyl-Beta-Cyclodextrin. J Assoc Res Otolaryngol 2015; 16:599-611. [PMID: 26055150 DOI: 10.1007/s10162-015-0528-6] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2015] [Accepted: 05/20/2015] [Indexed: 10/23/2022] Open
Abstract
Cyclodextrins are simple yet powerful molecules widely used in medicinal formulations and industry for their ability to stabilize and solubilize guest compounds. However, recent evidence shows that 2-hydroxypropyl-β-cyclodextrin (HPβCD) causes severe hearing loss in mice, selectively killing outer hair cells (OHC) within 1 week of subcutaneous drug treatment. In the current study, the impact of HPβCD on auditory physiology and pathology was explored further as a function of time and route of administration. When administered subcutaneously or directly into cerebrospinal fluid, single injections of HPβCD caused up to 60 dB threshold shifts and widespread OHC loss in a dose-dependent manner. Combined dosing caused no greater deficit, suggesting a common mode of action. After drug treatment, OHC loss progressed over time, beginning in the base and extending toward the apex, creating a sharp transition between normal and damaged regions of the cochlea. Administration into cerebrospinal fluid caused rapid ototoxicity when compared to subcutaneous delivery. Despite the devastating effect on the cochlea, HPβCD was relatively safe to other peripheral and central organ systems; specifically, it had no notable nephrotoxicity in contrast to other ototoxic compounds like aminoglycosides and platinum-based drugs. As cyclodextrins find expanding medicinal applications, caution should be exercised as these drugs possess a unique, poorly understood, ototoxic mechanism.
Collapse
|
49
|
Tatti M, Motta M, Scarpa S, Di Bartolomeo S, Cianfanelli V, Tartaglia M, Salvioli R. BCM-95 and (2-hydroxypropyl)-β-cyclodextrin reverse autophagy dysfunction and deplete stored lipids in Sap C-deficient fibroblasts. Hum Mol Genet 2015; 24:4198-211. [PMID: 25926625 DOI: 10.1093/hmg/ddv153] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Accepted: 04/26/2015] [Indexed: 12/19/2022] Open
Abstract
Saposin (Sap) C deficiency is a rare variant form of Gaucher disease caused by impaired Sap C expression or accelerated degradation, and associated with accumulation of glucosylceramide and other lipids in the endo/lysosomal compartment. No effective therapies are currently available for the treatment of Sap C deficiency. We previously reported that a reduced amount and enzymatic activity of cathepsin (Cath) B and Cath D, and defective autophagy occur in Sap C-deficient fibroblasts. Here, we explored the use of two compounds, BCM-95, a curcumin derivative, and (2-hydroxypropyl)-β-cyclodextrin (HP-β-CD), to improve lysosomal function of Sap C-deficient fibroblasts. Immunofluorescence and biochemical studies documented that each compound promotes an increase of the expression levels and activities of Cath B and Cath D, and efficient clearance of cholesterol (Chol) and ceramide (Cer) in lysosomes. We provide evidence that BCM-95 and HP-β-CD enhance lysosomal function promoting autophagic clearance capacity and lysosome reformation. Our findings suggest a novel pharmacological approach to Sap C deficiency directed to treat major secondary pathological aspects in this disorder.
Collapse
Affiliation(s)
- Massimo Tatti
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy
| | - Marialetizia Motta
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy
| | - Susanna Scarpa
- Department of Experimental Medicine, University of Rome 'La Sapienza', Viale Regina Elena, 324, 00161 Rome, Italy
| | - Sabrina Di Bartolomeo
- Dulbecco Telethon Institute at the Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy and
| | - Valentina Cianfanelli
- Dulbecco Telethon Institute at the Department of Biology, University of Rome Tor Vergata, Via della Ricerca Scientifica, 00133 Rome, Italy and Unit of Cell Stress and Survival, Danish Cancer Society Research Center, Strandboulevarden 49, 2100 Copenhagen, Denmark
| | - Marco Tartaglia
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy
| | - Rosa Salvioli
- Department of Haematology, Oncology and Molecular Medicine, Istituto Superiore di Sanità, Viale Regina Elena, 299, 00161 Rome, Italy,
| |
Collapse
|
50
|
Maeda Y, Motoyama K, Higashi T, Horikoshi Y, Takeo T, Nakagata N, Kurauchi Y, Katsuki H, Ishitsuka Y, Kondo Y, Irie T, Furuya H, Era T, Arima H. Effects of cyclodextrins on GM1-gangliosides in fibroblasts from GM1-gangliosidosis patients. J Pharm Pharmacol 2015; 67:1133-42. [PMID: 25851126 DOI: 10.1111/jphp.12405] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2014] [Accepted: 01/25/2015] [Indexed: 01/06/2023]
Abstract
OBJECTIVES GM1-gangliosidosis is an inherited disorder characterized by the accumulation of GM1-gangliosides in many tissues and organs, particularly in the brain. Currently, there is no treatment available for patients with ganglioside storage diseases. Therefore, we investigated the effects of cyclodextrins (CyDs) on the GM1-ganglioside level in EA1 cells, fibroblasts from patients with GM1-gangliosidosis. METHODS The concentrations of cholesterol and phospholipids in supernatants were determined by Cholesterol E-test Wako and Phospholipid C-test Wako, respectively. The effects of CyDs on GM1-ganglioside levels in EA1 cells using fluorescence-labelled cholera toxin B-subunit, which can bind to GM1-gangliosides specifically, were investigated by flow cytometry and confocal laser scanning microscopy. KEY FINDINGS The treatment with methylated CyDs, hydroxypropylated CyDs and branched CyDs decreased GM1-ganglioside levels in EA1 cells at 1 mm for 24 h. Unexpectedly, there was no significant change in the efflux of cholesterol or phospholipids from the cells after treatment with CyDs under the same experimental conditions, indicating that the efflux of membrane components is not associated with down-regulation of GM1-ganglioside levels in EA1 cells upon CyDs treatment. CONCLUSIONS CyDs may have the potential as drugs for GM1-gangliosidosis, although the mechanism should be thereafter clarified.
Collapse
Affiliation(s)
- Yuki Maeda
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.,Program for Leading Graduate Schools 'HIGO (Health Life Science: Interdisciplinary and Glocal Oriented) Program', Kumamoto University, Kumamoto, Japan
| | - Keiichi Motoyama
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Taishi Higashi
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuka Horikoshi
- Division of Reproductive Engineering, Center for Animal Resources and Development, Kumamoto University, Kumamoto, Japan
| | - Toru Takeo
- Division of Reproductive Engineering, Center for Animal Resources and Development, Kumamoto University, Kumamoto, Japan
| | - Naomi Nakagata
- Division of Reproductive Engineering, Center for Animal Resources and Development, Kumamoto University, Kumamoto, Japan
| | - Yuki Kurauchi
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.,Program for Leading Graduate Schools 'HIGO (Health Life Science: Interdisciplinary and Glocal Oriented) Program', Kumamoto University, Kumamoto, Japan
| | - Hiroshi Katsuki
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yoichi Ishitsuka
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Yuki Kondo
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan
| | - Tetsumi Irie
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.,Program for Leading Graduate Schools 'HIGO (Health Life Science: Interdisciplinary and Glocal Oriented) Program', Kumamoto University, Kumamoto, Japan
| | - Hirokazu Furuya
- Department of Neurology, Kochi Medical School, Kochi University, Kochi, Japan
| | - Takumi Era
- Department of Cell Modulation, Institute of Molecular Embryology and Genetics, Kumamoto University, Kumamoto, Japan
| | - Hidetoshi Arima
- Graduate School of Pharmaceutical Sciences, Kumamoto University, Kumamoto, Japan.,Program for Leading Graduate Schools 'HIGO (Health Life Science: Interdisciplinary and Glocal Oriented) Program', Kumamoto University, Kumamoto, Japan
| |
Collapse
|