51
|
Falcicchia C, Tozzi F, Arancio O, Watterson DM, Origlia N. Involvement of p38 MAPK in Synaptic Function and Dysfunction. Int J Mol Sci 2020; 21:ijms21165624. [PMID: 32781522 PMCID: PMC7460549 DOI: 10.3390/ijms21165624] [Citation(s) in RCA: 108] [Impact Index Per Article: 27.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2020] [Revised: 08/03/2020] [Accepted: 08/05/2020] [Indexed: 12/19/2022] Open
Abstract
Many studies have revealed a central role of p38 MAPK in neuronal plasticity and the regulation of long-term changes in synaptic efficacy, such as long-term potentiation (LTP) and long-term depression (LTD). However, p38 MAPK is classically known as a responsive element to stress stimuli, including neuroinflammation. Specific to the pathophysiology of Alzheimer’s disease (AD), several studies have shown that the p38 MAPK cascade is activated either in response to the Aβ peptide or in the presence of tauopathies. Here, we describe the role of p38 MAPK in the regulation of synaptic plasticity and its implication in an animal model of neurodegeneration. In particular, recent evidence suggests the p38 MAPK α isoform as a potential neurotherapeutic target, and specific inhibitors have been developed and have proven to be effective in ameliorating synaptic and memory deficits in AD mouse models.
Collapse
Affiliation(s)
- Chiara Falcicchia
- Institute of Neuroscience, Italian National Research Council, 56124 Pisa, Italy;
| | - Francesca Tozzi
- Bio@SNS laboratory, Scuola Normale Superiore, 56124 Pisa, Italy;
| | - Ottavio Arancio
- Taub Institute for Research on Alzheimer’s Disease and the Aging Brain, Columbia University, New York, NY 10032, USA;
| | | | - Nicola Origlia
- Institute of Neuroscience, Italian National Research Council, 56124 Pisa, Italy;
- Correspondence: ; Tel.: +39-050-3153193
| |
Collapse
|
52
|
Iannuzzi F, Sirabella R, Canu N, Maier TJ, Annunziato L, Matrone C. Fyn Tyrosine Kinase Elicits Amyloid Precursor Protein Tyr682 Phosphorylation in Neurons from Alzheimer's Disease Patients. Cells 2020; 9:E1807. [PMID: 32751526 PMCID: PMC7463977 DOI: 10.3390/cells9081807] [Citation(s) in RCA: 28] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/22/2020] [Revised: 07/23/2020] [Accepted: 07/27/2020] [Indexed: 12/22/2022] Open
Abstract
Alzheimer's disease (AD) is an incurable neurodegenerative disorder with a few early detection strategies. We previously proposed the amyloid precursor protein (APP) tyrosine 682 (Tyr682) residue as a valuable target for the development of new innovative pharmacologic or diagnostic interventions in AD. Indeed, when APP is phosphorylated at Tyr682, it is forced into acidic neuronal compartments where it is processed to generate neurotoxic amyloid β peptides. Of interest, Fyn tyrosine kinase (TK) interaction with APP Tyr682 residue increases in AD neurons. Here we proved that when Fyn TK was overexpressed it elicited APP Tyr682 phosphorylation in neurons from healthy donors and promoted the amyloidogenic APP processing with Aβ peptides accumulation and neuronal death. Phosphorylation of APP at Tyr (pAPP-Tyr) increased in neurons of AD patients and AD neurons that exhibited high pAPP-Tyr also had higher Fyn TK activity. Fyn TK inhibition abolished the pAPP-Tyr and reduced Aβ42 secretion in AD neurons. In addition, the multidomain adaptor protein Fe65 controlled the Fyn-mediated pAPP-Tyr, warranting the possibility of targeting the Fe65-APP-Fyn pathway to develop innovative strategies in AD. Altogether, these results strongly emphasize the relevance of focusing on pAPP Tyr682 either for diagnostic purposes, as an early biomarker of the disease, or for pharmacological targeting, using Fyn TKI.
Collapse
Affiliation(s)
- Filomena Iannuzzi
- Department of Biomedicine, Aarhus University, Aarhus C, 8000 Aarhus, Denmark;
| | - Rossana Sirabella
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples Federico II, 80131 Naples, Italy;
| | - Nadia Canu
- Department of System Medicine, University of Rome “Tor Vergata”, 00133 Rome, Italy;
- Institute of Biochemistry and Cell Biology, CNR, 00015 Monterotondo, Rome, Italy
| | - Thorsten J. Maier
- Paul-Ehrlich-Institut, (Federal Institute for Vaccines and Biomedicines), 63225 Langen, Germany;
| | - Lucio Annunziato
- SDN Research Institute Diagnostics and Nuclear (IRCCS SDN), Gianturco, 80131 Naples, Italy
| | - Carmela Matrone
- Division of Pharmacology, Department of Neuroscience, School of Medicine, University of Naples Federico II, 80131 Naples, Italy;
| |
Collapse
|
53
|
Turner RS, Hebron ML, Lawler A, Mundel EE, Yusuf N, Starr JN, Anjum M, Pagan F, Torres‐Yaghi Y, Shi W, Mulki S, Ferrante D, Matar S, Liu X, Esposito G, Berkowitz F, Jiang X, Ahn J, Moussa C. Nilotinib Effects on Safety, Tolerability, and Biomarkers in Alzheimer's Disease. Ann Neurol 2020; 88:183-194. [PMID: 32468646 PMCID: PMC7383852 DOI: 10.1002/ana.25775] [Citation(s) in RCA: 71] [Impact Index Per Article: 17.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2020] [Revised: 05/06/2020] [Accepted: 05/06/2020] [Indexed: 12/31/2022]
Abstract
OBJECTIVE Preclinical evidence with nilotinib, a US Food and Drug Administration (FDA)-approved drug for leukemia, indicates improvement in Alzheimer's disease phenotypes. We investigated whether nilotinib is safe, and detectable in cerebrospinal fluid, and alters biomarkers and clinical decline in Alzheimer's disease. METHODS This single-center, phase 2, randomized, double-blind, placebo-controlled study investigated the safety, tolerability, and pharmacokinetics of nilotinib, and measured biomarkers in participants with mild to moderate dementia due to Alzheimer's disease. The diagnosis was supported by cerebrospinal fluid or amyloid positron emission tomography biomarkers. Nilotinib 150 mg versus matching placebo was taken orally once daily for 26 weeks followed by nilotinib 300 mg versus placebo for another 26 weeks. RESULTS Of the 37 individuals enrolled, 27 were women and the mean (SD) age was 70.7 (6.48) years. Nilotinib was well-tolerated, although more adverse events, particularly mood swings, were noted with the 300 mg dose. In the nilotinib group, central nervous system (CNS) amyloid burden was significantly reduced in the frontal lobe compared to the placebo group. Cerebrospinal fluid Aβ40 was reduced at 6 months and Aβ42 was reduced at 12 months in the nilotinib group compared to the placebo. Hippocampal volume loss was attenuated (-27%) at 12 months and phospho-tau-181 was reduced at 6 months and 12 months in the nilotinib group. INTERPRETATION Nilotinib is safe and achieves pharmacologically relevant cerebrospinal fluid concentrations. Biomarkers of disease were altered in response to nilotinib treatment. These data support a larger, longer, multicenter study to determine the safety and efficacy of nilotinib in Alzheimer's disease. ANN NEUROL 2020 ANN NEUROL 2020;88:183-194.
Collapse
Affiliation(s)
- Raymond S. Turner
- Memory Disorders Program, Department of NeurologyGeorgetown University Medical CenterWashingtonDCUSA
| | - Michaeline L. Hebron
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of NeurologyGeorgetown University Medical CenterWashingtonDCUSA
| | - Abigail Lawler
- Movement Disorders Clinic, Department of NeurologyMedStar Georgetown University HospitalWashingtonDCUSA
| | - Elizabeth E. Mundel
- Movement Disorders Clinic, Department of NeurologyMedStar Georgetown University HospitalWashingtonDCUSA
| | - Nadia Yusuf
- Movement Disorders Clinic, Department of NeurologyMedStar Georgetown University HospitalWashingtonDCUSA
| | - J. Nathan Starr
- Movement Disorders Clinic, Department of NeurologyMedStar Georgetown University HospitalWashingtonDCUSA
| | - Muhammad Anjum
- Movement Disorders Clinic, Department of NeurologyMedStar Georgetown University HospitalWashingtonDCUSA
| | - Fernando Pagan
- Movement Disorders Clinic, Department of NeurologyMedStar Georgetown University HospitalWashingtonDCUSA
| | - Yasar Torres‐Yaghi
- Movement Disorders Clinic, Department of NeurologyMedStar Georgetown University HospitalWashingtonDCUSA
| | - Wangke Shi
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of NeurologyGeorgetown University Medical CenterWashingtonDCUSA
| | - Sanjana Mulki
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of NeurologyGeorgetown University Medical CenterWashingtonDCUSA
| | - Dalila Ferrante
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of NeurologyGeorgetown University Medical CenterWashingtonDCUSA
| | - Sara Matar
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of NeurologyGeorgetown University Medical CenterWashingtonDCUSA
| | - Xiaoguang Liu
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of NeurologyGeorgetown University Medical CenterWashingtonDCUSA
| | | | - Frank Berkowitz
- Department of RadiologyMedStar Georgetown HospitalWashingtonDCUSA
| | - Xiong Jiang
- Department of NeuroscienceGeorgetown University Medical CenterWashingtonDCUSA
| | - Jaeil Ahn
- Department of Biostatistics, Bioinformatics and BiomathematicsGeorgetown University Medical CenterWashingtonDCUSA
| | - Charbel Moussa
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of NeurologyGeorgetown University Medical CenterWashingtonDCUSA
| |
Collapse
|
54
|
De Masi C, Spitalieri P, Murdocca M, Novelli G, Sangiuolo F. Application of CRISPR/Cas9 to human-induced pluripotent stem cells: from gene editing to drug discovery. Hum Genomics 2020; 14:25. [PMID: 32591003 PMCID: PMC7318728 DOI: 10.1186/s40246-020-00276-2] [Citation(s) in RCA: 47] [Impact Index Per Article: 11.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 06/17/2020] [Indexed: 12/12/2022] Open
Abstract
Human-induced pluripotent stem cells (hiPSCs) and CRISPR/Cas9 gene editing system represent two instruments of basic and translational research, which both allow to acquire deep insight about the molecular bases of many diseases but also to develop pharmacological research.This review is focused to draw up the latest technique of gene editing applied on hiPSCs, exploiting some of the genetic manipulation directed to the discovery of innovative therapeutic strategies. There are many expediencies provided by the use of hiPSCs, which can represent a disease model clinically relevant and predictive, with a great potential if associated to CRISPR/Cas9 technology, a gene editing tool powered by ease and precision never seen before.Here, we describe the possible applications of CRISPR/Cas9 to hiPSCs: from drug development to drug screening and from gene therapy to the induction of the immunological response to specific virus infection, such as HIV and SARS-Cov-2.
Collapse
Affiliation(s)
- Claudia De Masi
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Paola Spitalieri
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Michela Murdocca
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Giuseppe Novelli
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy
| | - Federica Sangiuolo
- Department of Biomedicine and Prevention, University of Rome "Tor Vergata", Rome, Italy.
| |
Collapse
|
55
|
Maackiain Ameliorates 6-Hydroxydopamine and SNCA Pathologies by Modulating the PINK1/Parkin Pathway in Models of Parkinson's Disease in Caenorhabditis elegans and the SH-SY5Y Cell Line. Int J Mol Sci 2020; 21:ijms21124455. [PMID: 32585871 PMCID: PMC7352553 DOI: 10.3390/ijms21124455] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/18/2020] [Accepted: 06/21/2020] [Indexed: 02/07/2023] Open
Abstract
The movement disorder Parkinson's disease (PD) is the second most frequently diagnosed neurodegenerative disease, and is associated with aging, the environment, and genetic factors. The intracellular aggregation of α-synuclein and the loss of dopaminergic neurons in the substantia nigra pars compacta are the pathological hallmark of PD. At present, there is no successful treatment for PD. Maackiain (MK) is a flavonoid extracted from dried roots of Sophora flavescens Aiton. MK has emerged as a novel agent for PD treatment that acts by inhibiting monoamine oxidase B. In this study, we assessed the neuroprotective potential of MK in Caenorhabditis elegans and investigated possible mechanism of this neuroprotection in the human SH-SY5Y cell line. We found that MK significantly reduced dopaminergic neuron damage in 6-hydroxydopamine (6-OHDA)-exposed worms of the BZ555 strain, with corresponding improvements in food-sensing behavior and life-span. In transgenic worms of strain NL5901 treated with 0.25 mM MK, the accumulation of α-synuclein was diminished by 27% (p < 0.01) compared with that in untreated worms. Moreover, in worms and the SH-SY5Y cell line, we confirmed that the mechanism of MK-mediated protection against PD pathology may include blocking apoptosis, enhancing the ubiquitin-proteasome system, and augmenting autophagy by increasing PINK1/parkin expression. The use of small interfering RNA to downregulate parkin expression in vivo and in vitro could reverse the benefits of MK in PD models. MK may have considerable therapeutic applications in PD.
Collapse
|
56
|
Karim MR, Liao EE, Kim J, Meints J, Martinez HM, Pletnikova O, Troncoso JC, Lee MK. α-Synucleinopathy associated c-Abl activation causes p53-dependent autophagy impairment. Mol Neurodegener 2020; 15:27. [PMID: 32299471 PMCID: PMC7164361 DOI: 10.1186/s13024-020-00364-w] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2019] [Accepted: 02/13/2020] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND Studies link c-Abl activation with the accumulation of pathogenic α-synuclein (αS) and neurodegeneration in Parkinson's disease (PD). Currently, c-Abl, a tyrosine kinase activated by cellular stress, is thought to promote αS pathology by either directly phosphorylating αS or by causing autophagy deficits. METHODS αS overexpressing transgenic (Tg) mice were used in this study. A53T Tg mice that express high levels of human mutant A53TαS under the control of prion protein promoter. Two different approaches were used in this study. Natural aging and seeding model of synucleinopathy. In seeding model, intracortical/intrastriatal (IC/IS) stereotaxic injection of toxic lysates was done using tissue lysates from end-stage symptomatic mice. In this study, nilotinib and pifithrin-α was used as a c-Abl and p53 inhibitor, respectively. Both Tg and non-transgenic (nTg) mice from each group were subjected to nilotinib (10 mg/kg) or vehicle (DMSO) treatment. Frozen brain tissues from PD and control human cases were analyzed. In vitro cells study was implied for c-Abl/p53 genetic manipulation to uncover signal transduction. RESULTS Herein, we show that the pathologic effects of c-Abl in PD also involve activation of p53, as c-Abl activation in a transgenic mouse model of α-synucleinopathy (TgA53T) and human PD cases are associated with the increased p53 activation. Significantly, active p53 in TgA53T neurons accumulates in the cytosol, which may lead to inhibition of autophagy. Thus, we hypothesized that c-Abl-dependent p53 activation contributes to autophagy impairment in α-synucleinopathy. In support of the hypothesis, we show that c-Abl activation is sufficient to inhibit autophagy in p53-dependent manner. Moreover, inhibition of either c-Abl, using nilotinib, or p53, using pifithrin-α, was sufficient to increase autophagic flux in neuronal cells by inducing phosphorylation of AMP-activated kinase (AMPK), ULK1 activation, and down-regulation of mTORC1 signaling. Finally, we show that pharmacological attenuation of c-Abl activity by nilotinib treatment in the TgA53T mouse model reduces activation of p53, stimulates autophagy, decreases accumulation αS pathology, and delays disease onset. CONCLUSION Collectively, our data show that c-Abl activation by α-synucleinopathy causes p53 dependent autophagy deficits and both c-Abl and p53 represent therapeutic target for PD.
Collapse
Affiliation(s)
- Md. Razaul Karim
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55414 USA
| | - Elly E. Liao
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55414 USA
| | - Jaekwang Kim
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55414 USA
- Present Address: Department of Neural Development and Disease, Korea Brain Research Institute (KBRI), Daegu, 41068 South Korea
| | - Joyce Meints
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55414 USA
| | | | - Olga Pletnikova
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA
| | - Juan C. Troncoso
- Department of Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21287 USA
| | - Michael K. Lee
- Department of Neuroscience, University of Minnesota, Minneapolis, MN 55414 USA
- Institute for Translational Neuroscience, University of Minnesota, 2101 6th Street SE, Minneapolis, MN 55414 USA
| |
Collapse
|
57
|
Cai Q, Jeong YY. Mitophagy in Alzheimer's Disease and Other Age-Related Neurodegenerative Diseases. Cells 2020; 9:cells9010150. [PMID: 31936292 PMCID: PMC7017092 DOI: 10.3390/cells9010150] [Citation(s) in RCA: 143] [Impact Index Per Article: 35.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2019] [Revised: 01/03/2020] [Accepted: 01/05/2020] [Indexed: 02/07/2023] Open
Abstract
Mitochondrial dysfunction is a central aspect of aging and neurodegenerative diseases, including Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis, and Huntington's disease. Mitochondria are the main cellular energy powerhouses, supplying most of ATP by oxidative phosphorylation, which is required to fuel essential neuronal functions. Efficient removal of aged and dysfunctional mitochondria through mitophagy, a cargo-selective autophagy, is crucial for mitochondrial maintenance and neuronal health. Mechanistic studies into mitophagy have highlighted an integrated and elaborate cellular network that can regulate mitochondrial turnover. In this review, we provide an updated overview of the recent discoveries and advancements on the mitophagy pathways and discuss the molecular mechanisms underlying mitophagy defects in Alzheimer's disease and other age-related neurodegenerative diseases, as well as the therapeutic potential of mitophagy-enhancing strategies to combat these disorders.
Collapse
|
58
|
Zhao Y, Zhang Y, Zhang J, Zhang X, Yang G. Molecular Mechanism of Autophagy: Its Role in the Therapy of Alzheimer's Disease. Curr Neuropharmacol 2020; 18:720-739. [PMID: 31934838 PMCID: PMC7536828 DOI: 10.2174/1570159x18666200114163636] [Citation(s) in RCA: 16] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2019] [Revised: 11/04/2019] [Accepted: 01/11/2020] [Indexed: 12/12/2022] Open
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder of progressive dementia that is characterized by the accumulation of beta-amyloid (Aβ)-containing neuritic plaques and intracellular Tau protein tangles. This distinctive pathology indicates that the protein quality control is compromised in AD. Autophagy functions as a "neuronal housekeeper" that eliminates aberrant protein aggregates by wrapping then into autophagosomes and delivering them to lysosomes for degradation. Several studies have suggested that autophagy deficits in autophagy participate in the accumulation and propagation of misfolded proteins (including Aβ and Tau). In this review, we summarize current knowledge of autophagy in the pathogenesis of AD, as well as some pathways targeting the restoration of autophagy. Moreover, we discuss how these aspects can contribute to the development of disease-modifying therapies in AD.
Collapse
Affiliation(s)
| | | | | | | | - Guofeng Yang
- Address correspondence to this author at the Department of Geriatrics, Second Hospital of Hebei Medical University, 215 Hepingxi Road, Shijiazhuang, 050000, China; Tel: +86-311-66636243; E-mail:
| |
Collapse
|
59
|
Huang YF, Gu CJ, Wang Q, Xu L, Chen J, Zhou W, Zhou Z, Zhao SJ, Li LW, Kong FQ, Qian DF, Zhao X, Fan J, Li QQ, Yin GY. The protective effort of GPCR kinase 2-interacting protein-1 in neurons via promoting Beclin1-Parkin induced mitophagy at the early stage of spinal cord ischemia-reperfusion injury. FASEB J 2019; 34:2055-2074. [PMID: 31908016 DOI: 10.1096/fj.201902047r] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2019] [Revised: 10/26/2019] [Accepted: 11/06/2019] [Indexed: 12/27/2022]
Abstract
In spinal cord ischemia-reperfusion (I/R) injury, large amounts of reactive oxygen species can cause mitochondrial damage. Therefore, mitophagy acts as the main mechanism for removing damaged mitochondria and protects nerve cells. This study aimed to illustrate the important role of GPCR kinase 2-interacting protein-1 (GIT1) in mitophagy in vivo and in vitro. The level of mitophagy in the neurons of Git1 knockout mice was significantly reduced after ischemia-reperfusion. However, the overexpression of adeno-associated virus with Git1 promoted mitophagy and inhibited the apoptosis of neurons. GIT1 regulated the phosphorylation of Beclin-1 in Thr119, which could promote the translocation of Parkin to the mitochondrial outer membrane. This process was independent of PTEN-induced kinase 1 (PINK1), but it could not rescue the role in the absence of PINK1. Overall, GIT1 enhanced mitophagy and protected neurons against ischemia-reperfusion injury and, hence, might serve as a new research site for the protection of ischemia-reperfusion injury.
Collapse
Affiliation(s)
- Yi-Fan Huang
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Chang-Jiang Gu
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Qian Wang
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Lin Xu
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China.,Department of Orthopedics, Wuxi Xishan People's Hospital, Wuxi, P.R. China
| | - Jian Chen
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Wei Zhou
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Zheng Zhou
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Shu-Jie Zhao
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Lin-Wei Li
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Fan-Qi Kong
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Ding-Fei Qian
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Xuan Zhao
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Jin Fan
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Qing-Qing Li
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| | - Guo-Yong Yin
- Department of Orthopedics, the First Affiliated Hospital of Nanjing Medical University, Nanjing, P.R. China
| |
Collapse
|
60
|
Multikinase Abl/DDR/Src Inhibition Produces Optimal Effects for Tyrosine Kinase Inhibition in Neurodegeneration. Drugs R D 2019; 19:149-166. [PMID: 30919310 PMCID: PMC6544596 DOI: 10.1007/s40268-019-0266-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
Background and objectives Inhibition of Abelson (Abl) tyrosine kinase as a therapeutic target has been gaining attention in neurodegeneration. Post-mortem Alzheimer’s and Parkinson’s disease brains show that the levels of several other tyrosine kinases, including Discoidin Domain Receptors (DDR1/2) are elevated. Knockdown of these tyrosine kinases with shRNA reduces neurotoxic proteins, including alpha-synuclein, beta-amyloid and tau. Methods Direct profiling of the pharmacokinetics of multi-kinase inhibitors Nilotinib, Bosutinib, Bafetinib, Radotinib and LCB-03-0110 shows differential levels of brain penetration but the ability of these agents to reduce toxic proteins is independent of brain concentration and selectivity to Abl. Results Our results indicate that the effective dose of Nilotinib has the lowest plasma:brain ratio (1%) followed by Bosutinib and Radotinib (5%), Bafetinib (12%) and LCB-03-0110 (12%). However, similar doses of multi-kinase Abl/DDR inhibitor Nilotinib, DDR/Src inhibitor LCB-03-0110 and Abl/Src inhibitor Bosutinib were much more effective than the more selective Abl inhibitors Radotinib and Bafetinib. Taken together, these data suggest that a multi-kinase target that includes Abl and other tyrosine kinases (DDRs, and Src) may offer more advantages alleviating neurodegenerative pathologies than the absolute CNS drug concentration and selectivity to Abl. Conclusion DDRs and Src are other potential co-targets with Abl in neurodegeneration. Electronic supplementary material The online version of this article (10.1007/s40268-019-0266-z) contains supplementary material, which is available to authorized users.
Collapse
|
61
|
Imamura K, Izumi Y, Banno H, Uozumi R, Morita S, Egawa N, Ayaki T, Nagai M, Nishiyama K, Watanabe Y, Hanajima R, Oki R, Fujita K, Takahashi N, Ikeda T, Shimizu A, Morinaga A, Hirohashi T, Fujii Y, Takahashi R, Inoue H. Induced pluripotent stem cell-based Drug Repurposing for Amyotrophic lateral sclerosis Medicine (iDReAM) study: protocol for a phase I dose escalation study of bosutinib for amyotrophic lateral sclerosis patients. BMJ Open 2019; 9:e033131. [PMID: 31796494 PMCID: PMC7003406 DOI: 10.1136/bmjopen-2019-033131] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
INTRODUCTION Amyotrophic lateral sclerosis (ALS) is a progressive and severe neurodegenerative disease caused by motor neuron death. There have as yet been no fundamental curative medicines, and the development of a medicine for ALS is urgently required. Induced pluripotent stem cell (iPSC)-based drug repurposing identified an Src/c-Abl inhibitor, bosutinib, as a candidate molecular targeted therapy for ALS. The objectives of this study are to evaluate the safety and tolerability of bosutinib for the treatment of patients with ALS and to explore the efficacy of bosutinib on ALS. This study is the first clinical trial of administered bosutinib for patients with ALS. METHODS AND ANALYSIS An open-label, multicentre phase I dose escalation study has been designed. The study consists of a 12-week observation period, a 1-week transitional period, a 12-week study treatment period and a 4-week follow-up period. After completion of the transitional period, subjects whose total ALS Functional Rating Scale-Revised (ALSFRS-R) score decreased by 1-3 points during the 12-week observation period receive bosutinib for 12 weeks. Three to six patients with ALS are enrolled in each of the four bosutinib dose levels (100, 200, 300 or 400 mg/day) to evaluate the safety and tolerability under a 3+3 dose escalation study design. Dose escalation and maximum tolerated dose are determined by the safety assessment committee comprising oncologists/haematologists and neurologists based on the incidence of dose-limiting toxicity in the first 4 weeks of the treatment at each dose level. A recommended phase II dose is determined by the safety assessment committee on completion of the 12-week study treatment in all subjects at all dose levels. The efficacy of bosutinib is also evaluated exploratorily using ALS clinical scores and biomarkers. ETHICS AND DISSEMINATION This study received full ethical approval from the institutional review board of each participating site. The findings of the study will be disseminated in peer-reviewed journals and at scientific conferences. TRIAL REGISTRATION NUMBER UMIN000036295; Pre-results, JMA-IIA00419; Pre-results.
Collapse
Affiliation(s)
- Keiko Imamura
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Yuishin Izumi
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Haruhiko Banno
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| | - Ryuji Uozumi
- Department of Biomedical Statistics and Bioinformatics, Kyoto University, Kyoto, Japan
| | - Satoshi Morita
- Department of Biomedical Statistics and Bioinformatics, Kyoto University, Kyoto, Japan
| | - Naohiro Egawa
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Takashi Ayaki
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Makiko Nagai
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Kazutoshi Nishiyama
- Department of Neurology, Kitasato University School of Medicine, Sagamihara, Japan
| | - Yasuhiro Watanabe
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Ritsuko Hanajima
- Division of Neurology, Department of Brain and Neurosciences, Faculty of Medicine, Tottori University, Yonago, Japan
| | - Ryosuke Oki
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Koji Fujita
- Department of Neurology, Tokushima University Graduate School of Biomedical Sciences, Tokushima, Japan
| | - Naoto Takahashi
- Department of Hematology, Nephrology, and Rheumatology, Akita University Graduate School of Medicine, Akita, Japan
| | - Takafumi Ikeda
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | - Akira Shimizu
- Institute for Advancement of Clinical and Translational Science (iACT), Kyoto University Hospital, Kyoto, Japan
| | | | | | | | - Ryosuke Takahashi
- Department of Neurology, Graduate School of Medicine, Kyoto University, Kyoto, Japan
| | - Haruhisa Inoue
- Center for iPS Cell Research and Application (CiRA), Kyoto University, Kyoto, Japan
| |
Collapse
|
62
|
Lee G, Cummings J, Decourt B, Leverenz JB, Sabbagh MN. Clinical drug development for dementia with Lewy bodies: past and present. Expert Opin Investig Drugs 2019; 28:951-965. [PMID: 31614096 PMCID: PMC6823159 DOI: 10.1080/13543784.2019.1681398] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2019] [Accepted: 10/14/2019] [Indexed: 12/25/2022]
Abstract
Introduction: Dementia with Lewy bodies (DLB) is an under-researched area despite being the second most common type of degenerative dementia after Alzheimer's disease. It is an area of unmet need with no approved symptomatic or disease-modifying therapies. The pharmacological management of DLB is complex and challenging because early trials of drugs for DLB have resulted in no demonstrable efficacy. Randomized controlled trials (RCTs) in the DLB population have only recently been initiated. Understanding results from previous and current clinical trials in DLB can provide insights for future research and development.Areas covered: We provide an overview of the DLB drug development landscape and the current treatment strategies. We reviewed ClinicalTrials.gov to identify all clinical trials for the treatment of DLB.Expert opinion: DLB drug development has significantly improved in recent years with eight agents now in clinical trials. However, more rigorous RCTs are urgently needed. Diagnostic criteria must be optimized to accurately diagnose patients for clinical trials and care. New biomarker strategies are necessary to improve diagnostic capabilities and trial designs, and novel drug targets should be identified to develop DLB specific disease-modifying therapies. Evaluating the current drug development landscape can provide insight into how best to optimize development practices.
Collapse
Affiliation(s)
- Garam Lee
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - Jeffrey Cummings
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
- Department of Brain Health, School of Integrated Health Science, University of Nevada Las Vegas, Las Vegas, NV, USA
| | - Boris Decourt
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| | - James B Leverenz
- Cleveland Clinic Lou Ruvo Center for Brain Health, Cleveland, OH, USA
| | - Marwan N Sabbagh
- Cleveland Clinic Lou Ruvo Center for Brain Health, Las Vegas, NV, USA
| |
Collapse
|
63
|
Liu X, Hebron M, Shi W, Lonskaya I, Moussa CEH. Ubiquitin specific protease-13 independently regulates parkin ubiquitination and alpha-synuclein clearance in alpha-synucleinopathies. Hum Mol Genet 2019; 28:548-560. [PMID: 30329047 DOI: 10.1093/hmg/ddy365] [Citation(s) in RCA: 54] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2018] [Accepted: 10/09/2018] [Indexed: 12/22/2022] Open
Abstract
Ubiquitin specific proteases (USPs) are de-ubiquitinases (DUBs) that control protein ubiquitination cycle. The role of DUBs is poorly understood in neurodegenerative diseases. We found that USP13 is overexpressed in post-mortem Parkinson's disease (PD) brains. We investigated whether changes in USP13 levels can affect two molecules, parkin and alpha-synuclein, that are implicated in PD pathogenesis. Parkin is an E3 ubiquitin ligase that is regulated by ubiquitination and targets certain proteins for degradation, and alpha-synuclein may be ubiquitinated and recycled in the normal brain. We found that USP13 independently regulates parkin and alpha-synuclein ubiquitination in models of alpha-synucleinopathies. USP13 shRNA knockdown increases alpha-synuclein ubiquitination and clearance, in a parkin-independent manner. Furthermore, USP13 overexpression counteracts the effects of a tyrosine kinase inhibitor, Nilotinib, while USP13 knockdown facilitates Nilotinib effects on alpha-synculein clearance, suggesting that alpha-synuclein ubiquitnation is important for its clearance. These studies provide novel evidence of USP13 effects on parkin and alpha-synuclein metabolism and suggest that USP13 is a potential therapeutic target in the alpha-synucleinopathies.
Collapse
Affiliation(s)
- Xiaoguang Liu
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, National Parkinson's Foundation Center of Excellence, Lewy Body Dementia Research Center of Excellence, Georgetown University Medical Center, N.W. Washington D.C., USA
| | - Michaeline Hebron
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, National Parkinson's Foundation Center of Excellence, Lewy Body Dementia Research Center of Excellence, Georgetown University Medical Center, N.W. Washington D.C., USA
| | - Wangke Shi
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, National Parkinson's Foundation Center of Excellence, Lewy Body Dementia Research Center of Excellence, Georgetown University Medical Center, N.W. Washington D.C., USA
| | - Irina Lonskaya
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, National Parkinson's Foundation Center of Excellence, Lewy Body Dementia Research Center of Excellence, Georgetown University Medical Center, N.W. Washington D.C., USA
| | - Charbel E-H Moussa
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, National Parkinson's Foundation Center of Excellence, Lewy Body Dementia Research Center of Excellence, Georgetown University Medical Center, N.W. Washington D.C., USA
| |
Collapse
|
64
|
Mitochondrial Dysfunction in Alzheimer’s Disease and Progress in Mitochondria-Targeted Therapeutics. Curr Behav Neurosci Rep 2019. [DOI: 10.1007/s40473-019-00179-0] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
65
|
Ma S, Attarwala IY, Xie XQ. SQSTM1/p62: A Potential Target for Neurodegenerative Disease. ACS Chem Neurosci 2019; 10:2094-2114. [PMID: 30657305 DOI: 10.1021/acschemneuro.8b00516] [Citation(s) in RCA: 104] [Impact Index Per Article: 20.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Neurodegenerative diseases, characterized by a progressive loss of brain function, affect the lives of millions of individuals worldwide. The complexity of the brain poses a challenge for scientists trying to map the biochemical and physiological pathways to identify areas of pathological errors. Brain samples of patients with neurodegenerative diseases have been shown to contain large amounts of misfolded and abnormally aggregated proteins, resulting in dysfunction in certain brain centers. Removal of these abnormal molecules is essential in maintaining protein homeostasis and overall neuronal health. Macroautophagy is a major route by which cells achieve this. Administration of certain autophagy-enhancing compounds has been shown to provide therapeutic effects for individuals with neurodegenerative conditions. SQSTM1/p62 is a scaffold protein closely involved in the macroautophagy process. p62 functions to anchor the ubiquitinated proteins to the autophagosome membrane, promoting degradation of unwanted molecules. Modulators targeting p62 to induce autophagy and promote its protective pathways for aggregate protein clearance have high potential in the treatment of these conditions. Additionally, causal relationships have been found between errors in regulation of SQSTM1/p62 and the development of a variety of neurodegenerative disorders, including Alzheimer's, Parkinson's, Huntington's, amyotrophic lateral sclerosis, and frontotemporal lobar degeneration. Furthermore, SQSTM1/p62 also serves as a signaling hub for multiple pathways associated with neurodegeneration, providing a potential therapeutic target in the treatment of neurodegenerative diseases. However, rational design of a p62-oriented autophagy modulator that can balance the negative and positive functions of multiple domains in p62 requires further efforts in the exploration of the protein structure and pathological basis.
Collapse
Affiliation(s)
| | | | - Xiang-Qun Xie
- ID4Pharma LLC, Bridgeville, Pennsylvania 15017, United States
| |
Collapse
|
66
|
Pagan FL, Hebron ML, Wilmarth B, Torres‐Yaghi Y, Lawler A, Mundel EE, Yusuf N, Starr NJ, Arellano J, Howard HH, Peyton M, Matar S, Liu X, Fowler AJ, Schwartz SL, Ahn J, Moussa C. Pharmacokinetics and pharmacodynamics of a single dose Nilotinib in individuals with Parkinson's disease. Pharmacol Res Perspect 2019; 7:e00470. [PMID: 30906562 PMCID: PMC6412143 DOI: 10.1002/prp2.470] [Citation(s) in RCA: 63] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Revised: 02/07/2019] [Accepted: 02/07/2019] [Indexed: 01/17/2023] Open
Abstract
Nilotinib is a broad-based tyrosine kinase inhibitor with the highest affinity to inhibit Abelson (c-Abl) and discoidin domain receptors (DDR1/2). Preclinical evidence indicates that Nilotinib reduces the level of brain alpha-synuclein and attenuates inflammation in models of Parkinson's disease (PD). We previously showed that Nilotinib penetrates the blood-brain barrier (BBB) and potentially improves clinical outcomes in individuals with PD and dementia with Lewy bodies (DLB). We performed a physiologically based population pharmacokinetic/pharmacodynamic (popPK/PD) study to determine the effects of Nilotinib in a cohort of 75 PD participants. Participants were randomized (1:1:1:1:1) into five groups (n = 15) and received open-label random single dose (RSD) 150:200:300:400 mg Nilotinib vs placebo. Plasma and cerebrospinal fluid (CSF) were collected at 1, 2, 3, and 4 hours after Nilotinib administration. The results show that Nilotinib enters the brain in a dose-independent manner and 200 mg Nilotinib increases the level of 3,4-Dihydroxyphenylacetic acid (DOPAC) and homovanillic acid (HVA), suggesting alteration to dopamine metabolism. Nilotinib significantly reduces plasma total alpha-synuclein and appears to reduce CSF oligomeric: total alpha-synuclein ratio. Furthermore, Nilotinib significantly increases the CSF level of triggering receptors on myeloid cells (TREM)-2, suggesting an anti-inflammatory effect. Taken together, 200 mg Nilotinib appears to be an optimal single dose that concurrently reduces inflammation and engages surrogate disease biomarkers, including dopamine metabolism and alpha-synuclein.
Collapse
Affiliation(s)
- Fernando L. Pagan
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
- Movement Disorders ClinicDepartment of NeurologyMedStar Georgetown University HospitalWashingtonDistrict of Columbia
| | - Michaeline L. Hebron
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Barbara Wilmarth
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
- Movement Disorders ClinicDepartment of NeurologyMedStar Georgetown University HospitalWashingtonDistrict of Columbia
| | - Yasar Torres‐Yaghi
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
- Movement Disorders ClinicDepartment of NeurologyMedStar Georgetown University HospitalWashingtonDistrict of Columbia
| | - Abigail Lawler
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Elizabeth E. Mundel
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
- Movement Disorders ClinicDepartment of NeurologyMedStar Georgetown University HospitalWashingtonDistrict of Columbia
| | - Nadia Yusuf
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
- Movement Disorders ClinicDepartment of NeurologyMedStar Georgetown University HospitalWashingtonDistrict of Columbia
| | - Nathan J. Starr
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
- Movement Disorders ClinicDepartment of NeurologyMedStar Georgetown University HospitalWashingtonDistrict of Columbia
| | - Joy Arellano
- Movement Disorders ClinicDepartment of NeurologyMedStar Georgetown University HospitalWashingtonDistrict of Columbia
| | - Helen H. Howard
- Movement Disorders ClinicDepartment of NeurologyMedStar Georgetown University HospitalWashingtonDistrict of Columbia
| | - Margo Peyton
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Sara Matar
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Xiaoguang Liu
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Alan J. Fowler
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Sorell L. Schwartz
- Department of PharmacologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Jaeil Ahn
- Department of Biostatistics, Bioinformatics and BiomathematicsGeorgetown University Medical CenterWashingtonDistrict of Columbia
| | - Charbel Moussa
- Translational Neurotherapeutics ProgramLaboratory for Dementia and ParkinsonismDepartment of NeurologyGeorgetown University Medical CenterWashingtonDistrict of Columbia
| |
Collapse
|
67
|
Esteves AR, Filipe F, Magalhães JD, Silva DF, Cardoso SM. The Role of Beclin-1 Acetylation on Autophagic Flux in Alzheimer's Disease. Mol Neurobiol 2019; 56:5654-5670. [PMID: 30661206 DOI: 10.1007/s12035-019-1483-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2018] [Accepted: 01/10/2019] [Indexed: 01/25/2023]
Abstract
Macroautophagy impairment plays a key role in sporadic Alzheimer's disease (sAD) neurodegenerative process. Nevertheless, the mechanism(s) that lead to a deficiency in macroautophagy in AD remains elusive. In this work, we identify, for the first time that Beclin-1 acetylation status is implicated in the alterations in autophagy observed in AD neurodegeneration. We observed that Beclin-1 is deacetylated by sirtuin 1 (SIRT1) and acetylated by p300. In addition, Beclin-1 acetylation inhibits autophagosome maturation, leading to impairment in autophagic flux. We also analyzed some proteins known to be involved in the maturation of autophagosomes such as Rab7, which participates in the fusion step with lysosomes. We observed that increased expression of Rab7 might represent a response to boost the formation of large perinuclear lysosome clusters in accordance with an increase in lysosomal biogenesis determined by increase in LAMP-2A, LAMP-1, and cathepsin D expression in AD cells. Thus, our data provides strong evidences that Beclin-1 acetylation impairs the autophagic flux, and despite lysosomal biogenesis seems to be triggered as a compensatory response, autophagosome fusion with lysosomes is compromised contributing to AD neurodegeneration.
Collapse
Affiliation(s)
- A Raquel Esteves
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Filipa Filipe
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - João D Magalhães
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Diana F Silva
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - Sandra M Cardoso
- CNC-Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal.
- Institute of Cellular and Molecular Biology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal.
| |
Collapse
|
68
|
Abstract
The most common neurodegenerative diseases are Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease, frontotemporal lobar degeneration, and the motor neuron diseases, with AD affecting approximately 6% of people aged 65 years and older, and PD affecting approximately 1% of people aged over 60 years. Specific proteins are associated with these neurodegenerative diseases, as determined by both immunohistochemical studies on post-mortem tissue and genetic screening, where protein misfolding and aggregation are key hallmarks. Many of these proteins are shown to misfold and aggregate into soluble non-native oligomers and large insoluble protein deposits (fibrils and plaques), both of which may exert a toxic gain of function. Proteotoxicity has been examined intensively in cell culture and in in vivo models, and clinical trials of methods to attenuate proteotoxicity are relatively new. Therapies to enhance cellular protein quality control mechanisms such as upregulation of chaperones and clearance/degradation pathways, as well as immunotherapies against toxic protein conformations, are being actively pursued. In this article, we summarize the common pathophysiology of neurodegenerative disease, and review therapies in early-phase clinical trials that target the proteotoxic component of several neurodegenerative diseases.
Collapse
Affiliation(s)
- Luke McAlary
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, V6T 1Z1, Canada.
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, V6T 2B5, Canada.
| | - Steven S Plotkin
- Department of Physics and Astronomy, University of British Columbia, Vancouver, BC, V6T 1Z1, Canada.
- Genome Sciences and Technology Program, University of British Columbia, Vancouver, BC, V6T 1Z2, Canada.
| | - Neil R Cashman
- Djavad Mowafaghian Centre for Brain Health, University of British Columbia, Vancouver, BC, V6T 2B5, Canada.
| |
Collapse
|
69
|
Simonovitch S, Schmukler E, Masliah E, Pinkas-Kramarski R, Michaelson DM. The Effects of APOE4 on Mitochondrial Dynamics and Proteins in vivo. J Alzheimers Dis 2019; 70:861-875. [PMID: 31306119 PMCID: PMC7478177 DOI: 10.3233/jad-190074] [Citation(s) in RCA: 40] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
This study examined the effects of apolipoprotein E4 (APOE4), the most prevalent genetic risk factor for Alzheimer's disease (AD), on proteins involved in mitochondrial dynamics and autophagy, in the hippocampus of targeted replacement mice. Immunohistochemical measurements revealed that the levels of the mitochondrial fusion-mediating protein, MFN1, were higher, whereas those of corresponding fission-regulating protein, DRP-1, were lower in the hippocampus of ApoE4 mice than in the corresponding ApoE3 mice, indicating that APOE4 is associated with increased mitochondrial fusion and decreased fission. A similar ApoE4-driven decrease in DRP-1 was also observed in AD brains. The levels of the mitochondrial proteins COX1 and Tom40, were higher in the ApoE4 mice, which is consistent with the increased fusion. Measurements of the levels of cleaved PINK1 and parkin, which mark and target mitochondria for mitophagic degradation, revealed lower levels of cleaved PINK1, suggesting reduced mitochondrial membrane potential, and higher levels of parkin in the hippocampus of ApoE4 compared with the ApoE3 mice, indicating altered mitophagy. The levels of the ubiquitin-binding scaffold protein, p62/SQSTM1, which directs selected cargo to the autophagosomes, were also higher in the ApoE4 mice. These findings suggest that APOE4 is associated with enhanced mitochondrial fusion and decreased fission. Additionally, the results indicate that mitophagy/autophagy is reduced in ApoE4 mice, resulting in higher levels of proteins such as parkin and p62, which are normally degraded during this process. Taken together, these results suggest a novel mechanism that may underlie the pathological effects of APOE4 and indicate that use of APOE4 genotyping could pave the way for identification of novel APOE4-related therapeutic targets.
Collapse
Affiliation(s)
- Shira Simonovitch
- Department of Neurobiology, Sagol School of Neuroscience, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Eran Schmukler
- Department of Neurobiology, Sagol School of Neuroscience, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Eliezer Masliah
- Molecular Neuropathology Section, Laboratory of Neurogenetics, National Institute on Aging, National Institutes of Health, Bethesda, MD, USA
- Department Neurosciences, School of Medicine, University of California, La Jolla, San Diego, CA, USA
- Department of Pathology, School of Medicine, University of California, La Jolla, San Diego, CA, USA
| | - Ronit Pinkas-Kramarski
- Department of Neurobiology, Sagol School of Neuroscience, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Daniel M Michaelson
- Department of Neurobiology, Sagol School of Neuroscience, George S. Wise Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| |
Collapse
|
70
|
Liu X, Hebron ML, Mulki S, Wang C, Lekah E, Ferrante D, Shi W, Kurd-Misto B, Moussa C. Ubiquitin Specific Protease 13 Regulates Tau Accumulation and Clearance in Models of Alzheimer's Disease. J Alzheimers Dis 2019; 72:425-441. [PMID: 31594232 DOI: 10.3233/jad-190635] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Ubiquitin Specific Protease-13 (USP13) is a de-ubiquinating enzyme that regulates protein ubiquitination and clearance. The role of USP13 is largely unknown in neurodegeneration. In this study we aim to demonstrate whether tau accumulation and/or clearance depends on ubiquitination/de-ubiquitination via USP-13. We used transgenic animal models of human amyloid precursor protein (APP) or P301L tau mutations and genetically knocked-down USP13 expression via shRNA to determine USP13 effects on tau ubiquitination and levels. We found a two-fold increase of USP13 levels in postmortem Alzheimer's disease (AD) brains. USP13 knockdown significantly increased the activity of the 20S proteasome and reduced the levels of hyper-phosphorylated tau (p-tau) in primary cortical neurons. USP13 knockdown also reduced the levels of amyloid and increased p-tau ubiquitination and clearance in transgenic animal models that overexpress murine tau as a result of the expression of familial APP mutations (TgAPP) and the human mutant P301L tau (rTg4510), respectively. Clearance of p-tau appears to be mediated by autophagy in these animal models. Taken together, these data suggest that USP13 knockdown reduces p-tau accumulation via regulation of ubiquitination/de-ubiquitination and mediates its clearance via autophagy and/or the proteasome. These results suggest that USP13 inhibition may be a therapeutic strategy to reduce accumulation of plaques and toxic p-tau in AD and human tauopathies.
Collapse
Affiliation(s)
- Xiaoguang Liu
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Michaeline L Hebron
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Sanjana Mulki
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Chen Wang
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Elizabeth Lekah
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Dalila Ferrante
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Wangke Shi
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Bahjat Kurd-Misto
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| | - Charbel Moussa
- Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|
71
|
Abstract
KIT is a receptor tyrosine kinase that after binding to its ligand stem cell factor activates signaling cascades linked to biological processes such as proliferation, differentiation, migration and cell survival. Based on studies performed on SCF and/or KIT mutant animals that presented anemia, sterility, and/or pigmentation disorders, KIT signaling was mainly considered to be involved in the regulation of hematopoiesis, gametogenesis, and melanogenesis. More recently, novel animal models and ameliorated cellular and molecular techniques have led to the discovery of a widen repertoire of tissue compartments and functions that are being modulated by KIT. This is the case for the lung, heart, nervous system, gastrointestinal tract, pancreas, kidney, liver, and bone. For this reason, the tyrosine kinase inhibitors that were originally developed for the treatment of hemato-oncological diseases are being currently investigated for the treatment of non-oncological disorders such as asthma, rheumatoid arthritis, and alzheimer's disease, among others. The beneficial effects of some of these tyrosine kinase inhibitors have been proven to depend on KIT inhibition. This review will focus on KIT expression and regulation in healthy and pathologic conditions other than cancer. Moreover, advances in the development of anti-KIT therapies, including tyrosine kinase inhibitors, and their application will be discussed.
Collapse
|
72
|
Gwon Y, Kim SH, Kim HT, Kam TI, Park J, Lim B, Cha H, Chang HJ, Hong YR, Jung YK. Amelioration of amyloid β-FcγRIIb neurotoxicity and tau pathologies by targeting LYN. FASEB J 2018; 33:4300-4313. [PMID: 30540497 DOI: 10.1096/fj.201800926r] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022]
Abstract
SRC-family kinases (SFKs) have been implicated in Alzheimer's disease (AD), but their mode of action was scarcely understood. Here, we show that LYN plays an essential role in amyloid β (Aβ)-triggered neurotoxicity and tau hyperphosphorylation by phosphorylating Fcγ receptor IIb2 (FcγRIIb2). We found that enzyme activity of LYN was increased in the brain of AD patients and was promoted in neuronal cells exposed to Aβ 1-42 (Aβ1-42). Knockdown of LYN expression inhibited Aβ1-42-induced neuronal cell death. Of note, LYN interacted with FcγRIIb2 upon exposure to Aβ1-42 and phosphorylated FcγRIIb2 at Tyr273 within immunoreceptor tyrosine-based inhibitory motif in neuronal cells. With the use of the structure-based drug design, we isolated KICG2576, an ATP-competitive inhibitor of LYN. Determination of cocrystal structure illustrated that KICG2576 bound to the cleft in the LYN kinase domain and inhibited LYN with a half-maximal inhibitory concentration value of 0.15 μM. KICG2576 inhibited Aβ- or FcγRIIb2-induced cell death, and this effect was better than pyrazolopyrimidine 1, a widely used inhibitor of SFK. Upon exposure to Aβ, KICG2576 blocked the phosphorylation of FcγRIIb2 and translocation of phosphatidylinositol 3,4,5-trisphosphate 5-phosphatase 2, a binding protein to the phosphorylated FcγRIIb2, to the plasma membrane, resulting in the inhibition of tau hyperphosphorylation, the downstream event of Aβ1-42-FcγRIIb2 binding. Furthermore, intracerebroventricular injection of KICG2576 into mice ameliorated Aβ-induced memory impairment. These results suggest that LYN plays a crucial role in Aβ1-42-mediated neurotoxicity and tau pathology, providing a therapeutic potential of LYN in AD.-Gwon, Y., Kim, S.-H., Kim, H. T., Kam, T.-I., Park, J., Lim, B., Cha, H., Chang, H.-J., Hong, Y. R., Jung, Y.-K. Amelioration of amyloid β-FcγRIIb neurotoxicity and tau pathologies by targeting LYN.
Collapse
Affiliation(s)
- Youngdae Gwon
- School of Biological Sciences, Seoul National University, Seoul, South Korea; and
| | - Seo-Hyun Kim
- School of Biological Sciences, Seoul National University, Seoul, South Korea; and
| | - Hyun Tae Kim
- Crystalgenomics Incorporated, Gyeonggi-do, South Korea
| | - Tae-In Kam
- School of Biological Sciences, Seoul National University, Seoul, South Korea; and
| | - Jisu Park
- School of Biological Sciences, Seoul National University, Seoul, South Korea; and
| | - Bitna Lim
- School of Biological Sciences, Seoul National University, Seoul, South Korea; and
| | - Hyunju Cha
- Crystalgenomics Incorporated, Gyeonggi-do, South Korea
| | - Ho-Jin Chang
- Crystalgenomics Incorporated, Gyeonggi-do, South Korea
| | - Yong Rae Hong
- Crystalgenomics Incorporated, Gyeonggi-do, South Korea
| | - Yong-Keun Jung
- School of Biological Sciences, Seoul National University, Seoul, South Korea; and
| |
Collapse
|
73
|
Heard DS, Tuttle CSL, Lautenschlager NT, Maier AB. Repurposing Proteostasis-Modifying Drugs to Prevent or Treat Age-Related Dementia: A Systematic Review. Front Physiol 2018; 9:1520. [PMID: 30425653 PMCID: PMC6218672 DOI: 10.3389/fphys.2018.01520] [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: 07/30/2018] [Accepted: 10/09/2018] [Indexed: 12/21/2022] Open
Abstract
Background: Dementia has a significant impact on quality of life of older individuals. Impaired proteostasis has been implicated as a potential cause of dementia, that can be therapeutically targeted to improve patient outcomes. This review aimed to collate all current evidence of the potential for targeting proteostasis with repurposed drugs as an intervention for age-related dementia and cognitive decline. Methods: PubMed, Web of Science and Embase databases were searched from inception until 4th July 2017 for studies published in English. Interventional studies of repurposed proteostasis-modifying drugs in Alzheimer's disease (AD), Parkinson's disease (PD), Lewy Body disease, vascular dementia, and cognitive aging, in either animal models or humans with change in cognition as the outcome were included. The SYRCLE and Cochrane tools were used to assess risk of bias for included studies. Results: Overall 47 trials, 38 animal and 9 human, were isolated for inclusion in this review. Drugs tested in animals and humans included lithium, rapamycin, rifampicin, and tyrosine kinase inhibitors. Drugs tested only in animals included Macrophage and Granulocyte-Macrophage Colony Stimulating Factors, methylene blue, dantrolene, geranylgeranylacetone, minocycline and phenylbutyric acid. Lithium (n = 10 animal, n = 6 human) and rapamycin (n = 12 animal, n = 1 human) were the most studied proteostasis modifying drugs influencing cognition. Nine of ten animal studies of lithium showed a statistically significant benefit in Alzheimer's models. Rapamycin demonstrated a significant benefit in models of vascular dementia, aging, and Alzheimer's, but may not be effective in treating established Alzheimer's pathology. Lithium and nilotinib had positive outcomes in human studies including Alzheimer's and Parkinson's patients respectively, while a human study of rifampicin in Alzheimer's failed to demonstrate benefit. Microdose lithium showed a strongly significant benefit in both animals and humans. While the risk of bias was relatively low in human studies, the risk of bias in animal studies was largely unclear. Conclusion: Overall, the collective findings support the hypothesis that targeting proteostasis for treatment of dementia may be beneficial, and therefore future studies in humans with repurposed proteostasis modifying drugs are warranted. Larger human clinical trials focusing on safety, efficacy, tolerability, and reproducibility are required to translate these therapeutics into clinical practice.
Collapse
Affiliation(s)
- Daniel S Heard
- North West Mental Health, Melbourne Health, Melbourne, VIC, Australia
| | - Camilla S L Tuttle
- @AgeMelbourne, Department of Medicine and Aged Care, University of Melbourne, Melbourne, VIC, Australia
| | - Nicola T Lautenschlager
- North West Mental Health, Melbourne Health, Melbourne, VIC, Australia.,Academic Unit for Psychiatry of Old Age, Department of Psychiatry, University of Melbourne, Melbourne, VIC, Australia
| | - Andrea B Maier
- @AgeMelbourne, Department of Medicine and Aged Care, University of Melbourne, Melbourne, VIC, Australia.,@AgeAmsterdam, Department of Human Movement Sciences, Vrije Universiteit Amsterdam, Amsterdam Movement Sciences, Amsterdam, Netherlands
| |
Collapse
|
74
|
Briston T, Hicks AR. Mitochondrial dysfunction and neurodegenerative proteinopathies: mechanisms and prospects for therapeutic intervention. Biochem Soc Trans 2018; 46:829-842. [PMID: 29986938 PMCID: PMC6103456 DOI: 10.1042/bst20180025] [Citation(s) in RCA: 78] [Impact Index Per Article: 13.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2018] [Revised: 05/10/2018] [Accepted: 05/21/2018] [Indexed: 12/22/2022]
Abstract
Neurodegenerative proteinopathies are a group of pathologically similar, progressive disorders of the nervous system, characterised by structural alterations within and toxic misfolding of susceptible proteins. Oligomerisation of Aβ, tau, α-synuclein and TDP-43 leads to a toxin gain- or loss-of-function contributing to the phenotype observed in Alzheimer's disease, Parkinson's disease, amyotrophic lateral sclerosis and frontotemporal dementia. Misfolded proteins can adversely affect mitochondria, and post-mitotic neurones are especially sensitive to metabolic dysfunction. Misfolded proteins impair mitochondrial dynamics (morphology and trafficking), preventing functional mitochondria reaching the synapse, the primary site of ATP utilisation. Furthermore, a direct association of misfolded proteins with mitochondria may precipitate or augment dysfunctional oxidative phosphorylation and mitochondrial quality control, causing redox dyshomeostasis observed in disease. As such, a significant interest lies in understanding mechanisms of mitochondrial toxicity in neurodegenerative disorders and in dissecting these mechanisms with a view of maintaining mitochondrial homeostasis in disease. Recent advances in understanding mitochondrially controlled cell death pathways and elucidating the mitochondrial permeability pore bioarchitecture are beginning to present new avenues to target neurodegeneration. Novel mitochondrial roles of deubiquitinating enzymes are coming to light and present an opportunity for a new class of proteins to target therapeutically with the aim of promoting mitophagy and the ubiquitin-proteasome system. The brain is enormously metabolically active, placing a large emphasis on maintaining ATP supply. Therefore, identifying mechanisms to sustain mitochondrial function may represent a common intervention point across all proteinopathies.
Collapse
Affiliation(s)
- Thomas Briston
- Neurology Innovation Centre, Hatfield Research Laboratories, Eisai Ltd, Hatfield, U.K.
| | - Amy R Hicks
- Neurology Innovation Centre, Hatfield Research Laboratories, Eisai Ltd, Hatfield, U.K
| |
Collapse
|
75
|
Tau clearance improves astrocytic function and brain glutamate-glutamine cycle. J Neurol Sci 2018; 391:90-99. [PMID: 30103978 DOI: 10.1016/j.jns.2018.06.005] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 06/07/2018] [Accepted: 06/12/2018] [Indexed: 02/01/2023]
Abstract
Tau hyperphosphorylation is a critical factor in neurodegenerative diseases, including dementia and Parkinsonism. Existing animal models of tauopathies express tau in neurons within the forebrain and do not often show tau accumulation in the brainstem and astrocytes. This study aims to understand the effects of differential regional expression of tau on neurotransmitter balance in the brain. To obtain an animal model that expresses tau in the brainstem, we bred hemizygous mice that express P301L tau (TauP301L) and detected hyper-phosphorylated tau (p-tau) predominantly in the hippocampus, cortex, brainstem and thalamus. We previously demonstrated that TauP301L mice [26] express tau under the control of a prion promoter in both neurons and astrocytes, reminiscent of human tauopathies. We treated TauP301L mice with tyrosine kinase inhibitors (TKIs) to determine the effects of tau clearance on neurotransmitter balance and astrocytic function. 13C/1H MRS reveals astrocytic dysfunction via reduced glial aspartate and impaired glutamate-glutamine cycle. An increase in glutamate and GABA and decrease in glutamine were observed in homozygous mice compared to hemizygous and control littermates. Daily treatment with TKIs, nilotinib or bosutinib led to p-tau clearance via autophagy and reversal of neurotransmitter imbalance. These data suggest that accumulation of p-tau in the brainstem does not alter dopamine metabolism but may trigger glutamate toxicity and astrocytic dysfunction in the TauP301L mouse. TKIs reverse tau effects via reversal of neurotransmitter imbalance.
Collapse
|
76
|
Barbero-Camps E, Roca-Agujetas V, Bartolessis I, de Dios C, Fernández-Checa JC, Marí M, Morales A, Hartmann T, Colell A. Cholesterol impairs autophagy-mediated clearance of amyloid beta while promoting its secretion. Autophagy 2018; 14:1129-1154. [PMID: 29862881 PMCID: PMC6103708 DOI: 10.1080/15548627.2018.1438807] [Citation(s) in RCA: 81] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Macroautophagy/autophagy failure with the accumulation of autophagosomes is an early neuropathological feature of Alzheimer disease (AD) that directly affects amyloid beta (Aβ) metabolism. Although loss of presenilin 1 function has been reported to impair lysosomal function and prevent autophagy flux, the detailed mechanism leading to autophagy dysfunction in AD remains to be elucidated. The resemblance between pathological hallmarks of AD and Niemann-Pick Type C disease, including endosome-lysosome abnormalities and impaired autophagy, suggests cholesterol accumulation as a common link. Using a mouse model of AD (APP-PSEN1-SREBF2 mice), expressing chimeric mouse-human amyloid precursor protein with the familial Alzheimer Swedish mutation (APP695swe) and mutant presenilin 1 (PSEN1-dE9), together with a dominant-positive, truncated and active form of SREBF2/SREBP2 (sterol regulatory element binding factor 2), we demonstrated that high brain cholesterol enhanced autophagosome formation, but disrupted its fusion with endosomal-lysosomal vesicles. The combination of these alterations resulted in impaired degradation of Aβ and endogenous MAPT (microtubule associated protein tau), and stimulated autophagy-dependent Aβ secretion. Exacerbated Aβ-induced oxidative stress in APP-PSEN1-SREBF2 mice, due to cholesterol-mediated depletion of mitochondrial glutathione/mGSH, is critical for autophagy induction. In agreement, in vivo mitochondrial GSH recovery with GSH ethyl ester, inhibited autophagosome synthesis by preventing the oxidative inhibition of ATG4B deconjugation activity exerted by Aβ. Moreover, cholesterol-enrichment within the endosomes-lysosomes modified the levels and membrane distribution of RAB7A and SNAP receptors (SNAREs), which affected its fusogenic ability. Accordingly, in vivo treatment with 2-hydroxypropyl-β-cyclodextrin completely rescued these alterations, making it a potential therapeutic tool for AD.
Collapse
Affiliation(s)
- Elisabet Barbero-Camps
- a Department of Cell Death and Proliferation , Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain
| | - Vicente Roca-Agujetas
- a Department of Cell Death and Proliferation , Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain
| | - Isabel Bartolessis
- a Department of Cell Death and Proliferation , Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain
| | - Cristina de Dios
- a Department of Cell Death and Proliferation , Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain.,c Departament de Biomedicina, Facultat de Medicina , Universitat de Barcelona , Barcelona , Spain
| | - Jose C Fernández-Checa
- a Department of Cell Death and Proliferation , Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain.,d Liver Unit , Hospital Clinic, CIBEREHD , Barcelona , Spain , Research Center for Alcoholic Liver and Pancreatic Diseases , Keck School of Medicine of the University of Southern California , Los Angeles , CA , USA
| | - Montserrat Marí
- a Department of Cell Death and Proliferation , Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain
| | - Albert Morales
- a Department of Cell Death and Proliferation , Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain
| | - Tobias Hartmann
- e Experimental Neurology , Saarland University , Homburg/Saar , Germany
| | - Anna Colell
- a Department of Cell Death and Proliferation , Institut d'Investigacions Biomèdiques de Barcelona, Consejo Superior de Investigaciones Científicas (CSIC), Institut d'Investigacions Biomèdiques August Pi i Sunyer (IDIBAPS) , Barcelona , Spain.,b Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED) , Spain
| |
Collapse
|
77
|
Javidnia M, Hebron ML, Xin Y, Kinney NG, Moussa CEH. Pazopanib Reduces Phosphorylated Tau Levels and Alters Astrocytes in a Mouse Model of Tauopathy. J Alzheimers Dis 2018; 60:461-481. [PMID: 28869476 DOI: 10.3233/jad-170429] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Hyperphosphorylation and aggregation of tau protein is a critical factor in many neurodegenerative diseases. These diseases are increasing in prevalence, and there are currently no cures. Previous work from our group and others has shown that tyrosine kinase inhibitors (TKIs) can stimulate autophagy, decrease pathological proteins, and improve symptoms in models of neurodegeneration. Here we examined the role of pazopanib in mouse models that express either human mutant P301L tau (TauP301L) or triple mutant amyloid precursor protein (3x-AβPP). The TauP301L mouse expresses P301L tau under the control of a prion promoter in both neurons and astrocytes, reminiscent of some human tauopathies. Pazopanib crosses the blood-brain barrier with no detectable peripheral off-side effects, and decreases p-tau in TauP301L mice. Pazopanib reaches a brain concentration sufficient for inhibition of several tyrosine kinases, including vascular endothelial growth factor receptors (VEGFRs). Further, pazopanib does not affect microglia but reduces astrocyte levels toward nontransgenic controls in TauP301L mice. Pazopanib does not alter amyloid beta levels or astrocytes in 3x-AβPP mice but modulates a number of inflammatory markers (IP-10, MIP-1α, MIP-1β, and RANTES). These data suggest that pazopanib may be involved in p-tau clearance and modulation of astrocytic activity in models of tauopathies.
Collapse
Affiliation(s)
- Monica Javidnia
- Department of Neurology, Laboratory for Dementiaand Parkinsonism, Translational Neurotherapeutics Program, Washington, DC, USA.,Department of Pharmacologyand Physiology, Georgetown University Medical Center, Washington, DC, USA
| | - Michaeline L Hebron
- Department of Neurology, Laboratory for Dementiaand Parkinsonism, Translational Neurotherapeutics Program, Washington, DC, USA
| | - Yue Xin
- Department of Neurology, Laboratory for Dementiaand Parkinsonism, Translational Neurotherapeutics Program, Washington, DC, USA
| | - Nikolas G Kinney
- Department of Neurology, Laboratory for Dementiaand Parkinsonism, Translational Neurotherapeutics Program, Washington, DC, USA
| | - Charbel E-H Moussa
- Department of Neurology, Laboratory for Dementiaand Parkinsonism, Translational Neurotherapeutics Program, Washington, DC, USA
| |
Collapse
|
78
|
SESN2 facilitates mitophagy by helping Parkin translocation through ULK1 mediated Beclin1 phosphorylation. Sci Rep 2018; 8:615. [PMID: 29330382 PMCID: PMC5766514 DOI: 10.1038/s41598-017-19102-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/20/2017] [Accepted: 12/22/2017] [Indexed: 11/17/2022] Open
Abstract
Mitophagy, the selective degradation of mitochondria by autophagy, is crucial for the maintenance of healthy mitochondrial pool in cells. The critical event in mitophagy is the translocation of cytosolic Parkin, a ubiquitin ligase, to the surface of defective mitochondria. This study elucidates a novel role of SESN2/Sestrin2, a stress inducible protein, in mitochondrial translocation of PARK2/Parkin during mitophagy. The data demonstrates that SESN2 downregulation inhibits BECN1/Beclin1 and Parkin interaction, thereby preventing optimum mitochondrial accumulation of Parkin. SESN2 interacts with ULK1 (unc-51 like kinase 1) and assists ULK1 mediated phosphorylation of Beclin1 at serine-14 position required for binding with Parkin prior to mitochondrial translocation. The trigger for SESN2 activation and regulation of Parkin translocation is the generation of mitochondrial superoxide. Scavenging of mitochondrial superoxide lower the levels of SESN2, resulting in retardation of Parkin translocation. Importantly, we observe that SESN2 mediated cytosolic interaction of Parkin and Beclin1 is PINK1 independent but mitochondrial translocation of Parkin is PINK1 dependent. Together, these findings suggest the role of SESN2 as a positive regulator of Parkin mediated mitophagy.
Collapse
|
79
|
Abstract
Parkinson's disease is a progressive neurodegenerative disease characterized by Lewy body pathology of which the primary constituent is aggregated misfolded alpha-synuclein protein. Currently, there are no clinical therapies for treatment of the underlying alpha-synuclein dysfunction and accumulation, and the standard of care for patients with Parkinson's disease focuses only on symptom management, creating an immense therapeutic gap that needs to be filled. Defects in autophagy have been strongly implicated in Parkinson's disease. Here, we review evidence from human, mouse, and cell culture studies to briefly explain these defects in autophagy in Parkinson's disease and the necessity for autophagy to be carefully and precisely tuned to maintain neuron survival. We summarize recent experimental agents for treating alpha-synuclein accumulation in α-synuclein Parkinson's disease and related synucleinopathies. Most of the efforts for developing experimental agents have focused on immunotherapeutic strategies, but we discuss why those efforts are misplaced. Finally, we emphasize why increasing autophagy flux for alpha-synuclein clearance is the most promising therapeutic strategy. Activating autophagy has been successful in preclinical models of Parkinson's disease and yields promising results in clinical trials.
Collapse
Affiliation(s)
- Alan J Fowler
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Room 203-C, Building D, 4000 Reservoir Rd. NW, Washington, DC, USA
| | - Charbel E-H Moussa
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Translational Neurotherapeutics Program, Room 203-C, Building D, 4000 Reservoir Rd. NW, Washington, DC, USA.
| |
Collapse
|
80
|
Amanullah A, Upadhyay A, Joshi V, Mishra R, Jana NR, Mishra A. Progressing neurobiological strategies against proteostasis failure: Challenges in neurodegeneration. Prog Neurobiol 2017; 159:1-38. [DOI: 10.1016/j.pneurobio.2017.08.005] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2017] [Revised: 06/01/2017] [Accepted: 08/25/2017] [Indexed: 02/07/2023]
|
81
|
Li S, Le W. An insight review of autophagy biology and neurodegenerative diseases: machinery, mechanisms and regulation. SCIENCE CHINA-LIFE SCIENCES 2017; 60:1457-1459. [DOI: 10.1007/s11427-017-9214-7] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/11/2017] [Accepted: 11/21/2017] [Indexed: 10/18/2022]
|
82
|
Morel E, Mehrpour M, Botti J, Dupont N, Hamaï A, Nascimbeni AC, Codogno P. Autophagy: A Druggable Process. Annu Rev Pharmacol Toxicol 2017; 57:375-398. [PMID: 28061686 DOI: 10.1146/annurev-pharmtox-010716-104936] [Citation(s) in RCA: 113] [Impact Index Per Article: 16.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Macroautophagy (hereafter called autophagy) is a vacuolar, lysosomal pathway for catabolism of intracellular material that is conserved among eukaryotic cells. Autophagy plays a crucial role in tissue homeostasis, adaptation to stress situations, immune responses, and the regulation of the inflammatory response. Blockade or uncontrolled activation of autophagy is associated with cancer, diabetes, obesity, cardiovascular disease, neurodegenerative disease, autoimmune disease, infection, and chronic inflammatory disease. During the past decade, researchers have made major progress in understanding the three levels of regulation of autophagy in mammalian cells: signaling, autophagosome formation, and autophagosome maturation and lysosomal degradation. As we discuss in this review, each of these levels is potentially druggable, and, depending on the indication, may be able to stimulate or inhibit autophagy. We also summarize the different modulators of autophagy and their potential and limitations in the treatment of life-threatening diseases.
Collapse
Affiliation(s)
- Etienne Morel
- Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253, F-75993 Paris, France; .,Université Paris Descartes-Sorbonne Paris Cité, F-75012 Paris, France
| | - Maryam Mehrpour
- Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253, F-75993 Paris, France; .,Université Paris Descartes-Sorbonne Paris Cité, F-75012 Paris, France
| | - Joëlle Botti
- Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253, F-75993 Paris, France; .,Université Paris Diderot-Sorbonne Paris Cité, F-75993 Paris, France
| | - Nicolas Dupont
- Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253, F-75993 Paris, France; .,Université Paris Descartes-Sorbonne Paris Cité, F-75012 Paris, France
| | - Ahmed Hamaï
- Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253, F-75993 Paris, France; .,Université Paris Descartes-Sorbonne Paris Cité, F-75012 Paris, France
| | - Anna Chiara Nascimbeni
- Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253, F-75993 Paris, France; .,Université Paris Descartes-Sorbonne Paris Cité, F-75012 Paris, France
| | - Patrice Codogno
- Institut Necker-Enfants Malades (INEM), INSERM U1151-CNRS UMR 8253, F-75993 Paris, France; .,Université Paris Descartes-Sorbonne Paris Cité, F-75012 Paris, France
| |
Collapse
|
83
|
Assessment of Autophagy in Neurons and Brain Tissue. Cells 2017; 6:cells6030025. [PMID: 28832529 PMCID: PMC5617971 DOI: 10.3390/cells6030025] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2017] [Revised: 08/01/2017] [Accepted: 08/21/2017] [Indexed: 12/31/2022] Open
Abstract
Autophagy is a complex process that controls the transport of cytoplasmic components into lysosomes for degradation. This highly conserved proteolytic system involves dynamic and complex processes, using similar molecular elements and machinery from yeast to humans. Moreover, autophagic dysfunction may contribute to a broad spectrum of mammalian diseases. Indeed, in adult tissues, where the capacity for regeneration or cell division is low or absent (e.g., in the mammalian brain), the accumulation of proteins/peptides that would otherwise be recycled or destroyed may have pathological implications. Indeed, such changes are hallmarks of pathologies, like Alzheimer’s, Prion or Parkinson’s disease, known as proteinopathies. However, it is still unclear whether such dysfunction is a cause or an effect in these conditions. One advantage when analysing autophagy in the mammalian brain is that almost all the markers described in different cell lineages and systems appear to be present in the brain, and even in neurons. By contrast, the mixture of cell types present in the brain and the differentiation stage of such neurons, when compared with neurons in culture, make translating basic research to the clinic less straightforward. Thus, the purpose of this review is to describe and discuss the methods available to monitor autophagy in neurons and in the mammalian brain, a process that is not yet fully understood, focusing primarily on mammalian macroautophagy. We will describe some general features of neuronal autophagy that point to our focus on neuropathologies in which macroautophagy may be altered. Indeed, we centre this review around the hypothesis that enhanced autophagy may be able to provide therapeutic benefits in some brain pathologies, like Alzheimer’s disease, considering this pathology as one of the most prevalent proteinopathies.
Collapse
|
84
|
Discoidin domain receptor inhibition reduces neuropathology and attenuates inflammation in neurodegeneration models. J Neuroimmunol 2017; 311:1-9. [PMID: 28863860 DOI: 10.1016/j.jneuroim.2017.07.009] [Citation(s) in RCA: 38] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/07/2017] [Revised: 06/23/2017] [Accepted: 07/12/2017] [Indexed: 01/04/2023]
Abstract
The role of cell surface tyrosine kinase collagen-activated receptors known as discoidin domain receptors (DDRs) is unknown in neurodegenerative diseases. We detect up-regulation in DDRs level in post-mortem Alzheimer and Parkinson brains. Lentiviral shRNA knockdown of DDR1 and DDR2 reduces the levels of α-synuclein, tau, and β-amyloid and prevents cell loss in vivo and in vitro. DDR1 and DDR2 knockdown alters brain immunity and significantly reduces the level of triggering receptor expressed on myeloid cells (TREM)-2 and microglia. These studies suggest that DDR1 and DDR2 inhibition is a potential target to clear neurotoxic proteins and reduce inflammation in neurodegeneration.
Collapse
|
85
|
Heyburn L, Moussa CEH. TDP-43 in the spectrum of MND-FTLD pathologies. Mol Cell Neurosci 2017; 83:46-54. [PMID: 28687523 DOI: 10.1016/j.mcn.2017.07.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2017] [Revised: 06/28/2017] [Accepted: 07/04/2017] [Indexed: 12/12/2022] Open
Abstract
The relationship between RNA-binding proteins, particularly TAR DNA binding protein 43 (TDP-43), and neurodegeneration is an important area of research. TDP-43 is involved in so many cellular processes that perturbation of protein homeostasis can lead to countless downstream effects. Understanding what leads to this disease-related protein imbalance and the resulting cellular and molecular effects will help to develop targets for disease intervention, whether it be prevention of protein accumulation, or addressing a secondary effect of protein accumulation. Here we review the current literature of TDP-43 and TDP-43 pathologies, the effects of TDP-43 overexpression and disruption of synaptic proteins through its binding of messenger RNA, leading to synaptic dysfunction. This review highlights some of the still-limited knowledge of the protein TDP-43 and how it can contribute to disease.
Collapse
Affiliation(s)
- Lanier Heyburn
- Department of Neurology, Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Georgetown University Medical Center, Washington D.C. 20007, USA; Department of Pathology, Georgetown University Medical Center, Washington D.C., USA 20007.
| | - Charbel E-H Moussa
- Department of Neurology, Translational Neurotherapeutics Program, Laboratory for Dementia and Parkinsonism, Georgetown University Medical Center, Washington D.C. 20007, USA
| |
Collapse
|
86
|
Lee WJ, Moon J, Kim TJ, Jun JS, Lee HS, Ryu YJ, Lee ST, Jung KH, Park KI, Jung KY, Kim M, Lee SK, Chu K. The c-Abl inhibitor, nilotinib, as a potential therapeutic agent for chronic cerebellar ataxia. J Neuroimmunol 2017; 309:82-87. [PMID: 28601294 DOI: 10.1016/j.jneuroim.2017.05.015] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2017] [Revised: 05/16/2017] [Accepted: 05/21/2017] [Indexed: 10/19/2022]
Abstract
Nilotinib is a potent inhibitor of tyrosine kinase BCR-ABL that penetrates the blood-brain barrier. To evaluate the effect of nilotinib in chronic cerebellar ataxia, twelve patients with chronic cerebellar ataxia nonresponsive to other treatment options (modified Rankin scale [mRS] scores: >2) and received nilotinib therapy (daily doses: 150-300mg) for >4 (range 5-16) weeks were reviewed. At follow-up, improved mRS scores were found in 7/12 (58.3%) patients and favorable mRS scores (≤2) were found in 6/12 (50.0%) patients. No severe adverse event was observed. Atrophy in the cerebellar vermis appeared to be negatively associated with favorable outcomes.
Collapse
Affiliation(s)
- Woo-Jin Lee
- Department of Neurology, Comprehensive Epilepsy Center, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Jangsup Moon
- Department of Neurology, Comprehensive Epilepsy Center, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea; Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University, Seoul, South Korea
| | - Tae-Joon Kim
- Department of Neurology, Comprehensive Epilepsy Center, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea; Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University, Seoul, South Korea
| | - Jin-Sun Jun
- Department of Neurology, Comprehensive Epilepsy Center, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea; Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University, Seoul, South Korea
| | - Han Sang Lee
- Department of Neurology, Comprehensive Epilepsy Center, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea
| | - Young Jin Ryu
- Department of Radiology, Seoul National University Hospital, Seoul, South Korea
| | - Soon-Tae Lee
- Department of Neurology, Comprehensive Epilepsy Center, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea; Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University, Seoul, South Korea
| | - Keun-Hwa Jung
- Department of Neurology, Comprehensive Epilepsy Center, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea; Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University, Seoul, South Korea
| | - Kyung-Il Park
- Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University, Seoul, South Korea; Department of Neurology, Seoul National University Healthcare System Gangnam Center, Seoul, South Korea
| | - Ki-Young Jung
- Department of Neurology, Comprehensive Epilepsy Center, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea; Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University, Seoul, South Korea
| | - Manho Kim
- Department of Neurology, Comprehensive Epilepsy Center, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea; Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University, Seoul, South Korea; Protein Metabolism Research Center, Seoul National University College of Medicine, Seoul, South Korea
| | - Sang Kun Lee
- Department of Neurology, Comprehensive Epilepsy Center, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea; Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University, Seoul, South Korea
| | - Kon Chu
- Department of Neurology, Comprehensive Epilepsy Center, Laboratory for Neurotherapeutics, Biomedical Research Institute, Seoul National University Hospital, Seoul, South Korea; Program in Neuroscience, Neuroscience Research Institute of SNUMRC, College of Medicine, Seoul National University, Seoul, South Korea.
| |
Collapse
|
87
|
Khan SH, Zhao D, Shah SZA, Hassan MF, Zhu T, Song Z, Zhou X, Yang L. Parkin Overexpression Ameliorates PrP106-126-Induced Neurotoxicity via Enhanced Autophagy in N2a Cells. Cell Mol Neurobiol 2017; 37:717-728. [PMID: 27430567 DOI: 10.1007/s10571-016-0407-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2016] [Accepted: 07/12/2016] [Indexed: 10/21/2022]
Abstract
Transmissible spongiform encephalopathies (TSEs) are caused by the accumulation of the abnormal prion protein scrapie (PrPSc). Prion protein aggregation, misfolding, and cytotoxicity in the brain are the major causes of neuronal dysfunction and ultimate neurodegeneration in all TSEs. Parkin, an E3 ubiquitin ligase, has been studied extensively in all major protein misfolding aggregating diseases, especially Parkinson's disease and Alzheimer's disease, but the role of parkin in TSEs remains unknown. Here we investigated the role of parkin in a prion disease cell model in which neuroblastoma2a (N2a) cells were treated with prion peptide PrP106-126. We observed a gradual decrease in the soluble parkin level upon treatment with PrP106-126 in a time-dependent manner. Furthermore, endogenous parkin colocalized with FITC-tagged prion fragment106-126. Overexpression of parkin in N2a cells via transfection repressed apoptosis by enhancing autophagy. Parkin-overexpressing cells also showed reductions in apoptotic BAX translocation to the mitochondria and cytochrome c release to the cytosol, which ultimately inhibited activation of proapoptotic caspases. Taken together, our findings reveal a parkin-mediated cytoprotective mechanism against PrP106-126 toxicity, which is a novel potential therapeutic target for treating prion diseases.
Collapse
Affiliation(s)
- Sher Hayat Khan
- National Animal Transmissible Spongiform Encephalopathy Laboratory, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Deming Zhao
- National Animal Transmissible Spongiform Encephalopathy Laboratory, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Syed Zahid Ali Shah
- National Animal Transmissible Spongiform Encephalopathy Laboratory, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Mohammad Farooque Hassan
- National Animal Transmissible Spongiform Encephalopathy Laboratory, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Ting Zhu
- Fujian Agriculture and Forestry University, Fuzhou, China
| | - Zhiqi Song
- National Animal Transmissible Spongiform Encephalopathy Laboratory, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Xiangmei Zhou
- National Animal Transmissible Spongiform Encephalopathy Laboratory, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China
| | - Lifeng Yang
- National Animal Transmissible Spongiform Encephalopathy Laboratory, Key Laboratory of Animal Epidemiology and Zoonosis of Ministry of Agriculture, College of Veterinary Medicine and State Key Laboratory of Agrobiotechnology, China Agricultural University, Beijing, China.
| |
Collapse
|
88
|
Gelmetti V, De Rosa P, Torosantucci L, Marini ES, Romagnoli A, Di Rienzo M, Arena G, Vignone D, Fimia GM, Valente EM. PINK1 and BECN1 relocalize at mitochondria-associated membranes during mitophagy and promote ER-mitochondria tethering and autophagosome formation. Autophagy 2017; 13:654-669. [PMID: 28368777 DOI: 10.1080/15548627.2016.1277309] [Citation(s) in RCA: 238] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
Mitophagy is a highly specialized process to remove dysfunctional or superfluous mitochondria through the macroautophagy/autophagy pathway, aimed at protecting cells from the damage of disordered mitochondrial metabolism and apoptosis induction. PINK1, a neuroprotective protein mutated in autosomal recessive Parkinson disease, has been implicated in the activation of mitophagy by selectively accumulating on depolarized mitochondria, and promoting PARK2/Parkin translocation to them. While these steps have been characterized in depth, less is known about the process and site of autophagosome formation upon mitophagic stimuli. A previous study reported that, in starvation-induced autophagy, the proautophagic protein BECN1/Beclin1 (which we previously showed to interact with PINK1) relocalizes at specific regions of contact between the endoplasmic reticulum (ER) and mitochondria called mitochondria-associated membranes (MAM), from which the autophagosome originates. Here we show that, following mitophagic stimuli, autophagosomes also form at MAM; moreover, endogenous PINK1 and BECN1 were both found to relocalize at MAM, where they promoted the enhancement of ER-mitochondria contact sites and the formation of omegasomes, that represent autophagosome precursors. PARK2 was also enhanced at MAM following mitophagy induction. However, PINK1 silencing impaired BECN1 enrichment at MAM independently of PARK2, suggesting a novel role for PINK1 in regulating mitophagy. MAM have been recently implicated in many key cellular events. In this light, the observed prevalent localization of PINK1 at MAM may well explain other neuroprotective activities of this protein, such as modulation of mitochondrial calcium levels, mitochondrial dynamics, and apoptosis.
Collapse
Affiliation(s)
- Vania Gelmetti
- a Neurogenetics Unit, IRCCS Santa Lucia Foundation , Rome , Italy
| | - Priscilla De Rosa
- b IRCCS Casa Sollievo della Sofferenza, CSS-Mendel Institute , Rome , Italy
| | | | | | - Alessandra Romagnoli
- d National Institute for Infectious Diseases "LazzaroSpallanzani" IRCCS , Rome , Italy
| | - Martina Di Rienzo
- d National Institute for Infectious Diseases "LazzaroSpallanzani" IRCCS , Rome , Italy.,e Department of Biology , "Tor Vergata" University , Rome , Italy
| | - Giuseppe Arena
- f IRCM, Institut de Recherche en Cancérologie de Montpellier, INSERM U1194, Université Montpellier, Institutrégional du Cancer Montpellier , Montpellier , France
| | | | - Gian Maria Fimia
- d National Institute for Infectious Diseases "LazzaroSpallanzani" IRCCS , Rome , Italy.,h Department of Biological and Environmental Sciences and Technologies (DiSTeBA) , University of Salento , Lecce , Italy
| | - Enza Maria Valente
- a Neurogenetics Unit, IRCCS Santa Lucia Foundation , Rome , Italy.,i Department of Molecular Medicine , University of Pavia , Pavia , Italy
| |
Collapse
|
89
|
Castoria G, Auricchio F, Migliaccio A. Extranuclear partners of androgen receptor: at the crossroads of proliferation, migration, and neuritogenesis. FASEB J 2016; 31:1289-1300. [PMID: 28031322 DOI: 10.1096/fj.201601047r] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2016] [Accepted: 12/19/2016] [Indexed: 01/11/2023]
Abstract
In this review, we focus on the role played by the protein partners of ligand-activated extranuclear androgen receptor (AR) in the final effects of hormone action, such as proliferation, migration, and neuritogenesis. The choice of AR partner, at least in part, depends on cell type. Androgen-activated receptor directly associates with cytoplasmic Src tyrosine kinase in epithelial cells, whereas in mesenchymal and neuronal cells, it prevalently interacts with filamin A. In the former, proliferation represents the final hormonal outcome, whereas in the latter, either migration or neuritogenesis, respectively, occurs. Furthermore, AR partner filamin A is replaced with Src when mesenchymal cells are stimulated with very low androgen concentrations. Consequently, the migratory effect is replaced by mitogenesis. Use of peptides that prevent receptor/partner assembly abolishes the effects that are dependent on their association and offers new therapeutic approaches to AR-related diseases. Perturbation of migration is often associated with metastatic spreading in cancer. In turn, cell cycle aberration causes tumors to grow faster, whereas toxic signaling triggers neurodegenerative events in the CNS. Here, we provide examples of new tools that interfere in rapid androgen effects, including migration, proliferation, and neuronal differentiation, together with their potential therapeutic applications in AR-dependent diseases-mainly prostate cancer and neurodegenerative disorders.-Castoria, G., Auricchio, F., Migliaccio, A. Extranuclear partners of androgen receptor: at the crossroads of proliferation, migration, and neuritogenesis.
Collapse
Affiliation(s)
- Gabriella Castoria
- Department of Biochemistry, Biophysics, and General Pathology, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Ferdinando Auricchio
- Department of Biochemistry, Biophysics, and General Pathology, University of Campania "Luigi Vanvitelli," Naples, Italy
| | - Antimo Migliaccio
- Department of Biochemistry, Biophysics, and General Pathology, University of Campania "Luigi Vanvitelli," Naples, Italy
| |
Collapse
|
90
|
Abstract
The Abelson tyrosine kinases were initially identified as drivers of leukemia in mice and humans. The Abl family kinases Abl1 and Abl2 regulate diverse cellular processes during development and normal homeostasis, and their functions are subverted during inflammation, cancer and other pathologies. Abl kinases can be activated by multiple stimuli leading to cytoskeletal reorganization required for cell morphogenesis, motility, adhesion and polarity. Depending on the cellular context, Abl kinases regulate cell survival and proliferation. Emerging data support important roles for Abl kinases in pathologies linked to inflammation. Among these are neurodegenerative diseases and inflammatory pathologies. Unexpectedly, Abl kinases have also been identified as important players in mammalian host cells during microbial pathogenesis. Thus, the use of Abl kinase inhibitors might prove to be effective in the treatment of pathologies beyond leukemia and solid tumors. In this Cell Science at a Glance article and in the accompanying poster, we highlight the emerging roles of Abl kinases in the regulation of cellular processes in normal cells and diverse pathologies ranging from cancer to microbial pathogenesis.
Collapse
Affiliation(s)
- Aaditya Khatri
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Jun Wang
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| | - Ann Marie Pendergast
- Department of Pharmacology & Cancer Biology, Duke University School of Medicine, Durham, NC 27710, USA
| |
Collapse
|
91
|
Heyburn L, Hebron ML, Smith J, Winston C, Bechara J, Li Z, Lonskaya I, Burns MP, Harris BT, Moussa CEH. Tyrosine kinase inhibition reverses TDP-43 effects on synaptic protein expression, astrocytic function and amino acid dis-homeostasis. J Neurochem 2016; 139:610-623. [PMID: 27507246 DOI: 10.1111/jnc.13763] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2016] [Revised: 07/22/2016] [Accepted: 08/02/2016] [Indexed: 01/25/2023]
Abstract
The trans-activating response of DNA/RNA-binding protein (TDP)-43 pathology is associated with many neurodegenerative diseases via unknown mechanisms. Here, we use a transgenic mouse model over-expressing human wild-type neuronal TDP-43 to study the effects of TDP-43 pathology on glutamate metabolism and synaptic function. We found that neuronal TDP-43 over-expression affects synaptic protein expression, including Synapsin I, and alters surrounding astrocytic function. TDP-43 over-expression is associated with an increase in glutamate and γ-amino butyric acid and reduction of glutamine and aspartate levels, indicating impairment of presynaptic terminal. TDP-43 also decreases tricarboxylic acid cycle metabolism and induces oxidative stress via lactate accumulation. Neuronal TDP-43 does not alter microglia activity or significantly changes systemic and brain inflammatory markers compared to control. We previously demonstrated that brain-penetrant tyrosine kinase inhibitors (TKIs), nilotinib and bosutinib, reduce TDP-43-induced cell death in transgenic mice. Here, we show that TKIs reverse the effects of TDP-43 on synaptic proteins, increase astrocytic function and restore glutamate and neurotransmitter balance in TDP-43 mice. Nilotinib, but not bosutinib, reverses mitochondrial impairment and oxidative metabolism. Taken together, these data suggest that TKIs can attenuate TDP-43 toxicity and improve synaptic and astrocytic function, independent of microglial or other inflammatory effects. In conclusion, our data demonstrate novel mechanisms of the effects of neuronal TDP-43 over-expression on synaptic protein expression and alteration of astrocytic function.
Collapse
Affiliation(s)
- Lanier Heyburn
- Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, District of Columbia, USA.,Department of Pathology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Michaeline L Hebron
- Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Jacqueline Smith
- Drug Discovery Center, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Charisse Winston
- Trauma and Dementia Laboratory, Department of Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - John Bechara
- Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Zhaoxia Li
- Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, District of Columbia, USA.,School of Traditional Chinese Medicine, Capital Medical University, Fengtai District, Beijing, China
| | - Irina Lonskaya
- Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Mark P Burns
- Trauma and Dementia Laboratory, Department of Neuroscience, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Brent T Harris
- Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, District of Columbia, USA.,Department of Pathology, Georgetown University Medical Center, Washington, District of Columbia, USA
| | - Charbel E-H Moussa
- Laboratory for Dementia and Parkinsonism, Department of Neurology, Georgetown University Medical Center, Washington, District of Columbia, USA
| |
Collapse
|
92
|
Nishioka H, Tooi N, Isobe T, Nakatsuji N, Aiba K. BMS-708163 and Nilotinib restore synaptic dysfunction in human embryonic stem cell-derived Alzheimer's disease models. Sci Rep 2016; 6:33427. [PMID: 27641902 PMCID: PMC5027582 DOI: 10.1038/srep33427] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2016] [Accepted: 08/26/2016] [Indexed: 12/25/2022] Open
Abstract
Alzheimer’s disease (AD) is the most common form of dementia. Cellular AD models derived from human pluripotent stem cells are promising tools in AD research. We recently developed human embryonic stem cell-derived AD models which overexpress mutant Presenilin1 genes, and which exhibit AD phenotypes, including synaptic dysfunction. In this study, we found that our AD models showed reduced levels of RAB3A and SV2B proteins in the pre-synapses, which is a possible cause of electrophysiological abnormalities. Through the screening of chemical compounds using our AD models, we have identified Aβ peptide inhibitors which decrease the concentration of Aβ in culture supernatant. Among these, BMS-708163 and Nilotinib were found to improve the expression levels of RAB3A and SV2B proteins and to recover the electrophysiological function in our AD models. These results suggest that the AD models we developed are promising materials for the discovery of AD drugs that target the expression of pre-synaptic proteins and synaptic function.
Collapse
Affiliation(s)
- Hisae Nishioka
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan.,Graduate School of Medicine, Kyoto University, Kyoto 606-8501, Japan
| | - Norie Tooi
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Takehisa Isobe
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| | - Norio Nakatsuji
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan.,Institute for Frontier Medical Sciences, Kyoto University, Kyoto 606-8507, Japan
| | - Kazuhiro Aiba
- Institute for Integrated Cell-Material Sciences (WPI-iCeMS), Kyoto University, Kyoto 606-8501, Japan
| |
Collapse
|
93
|
Pagan F, Hebron M, Valadez EH, Torres-Yaghi Y, Huang X, Mills RR, Wilmarth BM, Howard H, Dunn C, Carlson A, Lawler A, Rogers SL, Falconer RA, Ahn J, Li Z, Moussa C. Nilotinib Effects in Parkinson's disease and Dementia with Lewy bodies. JOURNAL OF PARKINSON'S DISEASE 2016; 6:503-17. [PMID: 27434297 PMCID: PMC5008228 DOI: 10.3233/jpd-160867] [Citation(s) in RCA: 171] [Impact Index Per Article: 21.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND We evaluated the effects of low doses of the tyrosine kinase Abelson (Abl) inhibitor Nilotinib, on safety and pharmacokinetics in Parkinson's disease dementia or dementia with Lewy bodies. OBJECTIVES The primary outcomes of this study were safety and tolerability; pharmacokinetics and target engagement were secondary, while clinical outcomes were exploratory. METHODS Twelve subjects were randomized into 150 mg (n = 5) or 300 mg (n = 7) groups and received Nilotinib orally every day for 24 weeks. RESULTS This study shows that 150 mg and 300 mg doses of Nilotinib appear to be safe and tolerated in subjects with advanced Parkinson's disease. Nilotinib is detectable in the cerebrospinal fluid (CSF) and seems to engage the target Abl. Motor and cognitive outcomes suggest a possible beneficial effect on clinical outcomes. The CSF levels of homovanillic acid are significantly increased between baseline and 24 weeks of treatment. Exploratory CSF biomarkers were measured. CONCLUSIONS This small proof-of-concept study lacks a placebo group and participants were not homogenous, resulting in baseline differences between and within groups. This limits the interpretations of the biomarker and clinical data, and any conclusions should be drawn cautiously. Nonetheless, the collective observations suggest that it is warranted to evaluate the safety and efficacy of Nilotinib in larger randomized, double-blind, placebo-controlled trials.
Collapse
Affiliation(s)
- Fernando Pagan
- Department of Neurology, National Parkinson's Foundation Center for Excellence, Translational Neurotherapeutics Program. Movement Disorders Program. MedStar Georgetown Hospital Washington, DC, USA
| | - Michaeline Hebron
- Department of Neurology, Laboratory for Dementia and Parkinsonism, National Parkinson's Foundation Center for Excellence, Translational Neurotherapeutics Program. Georgetown University Medical Center, Washington, DC, USA
| | - Ellen H Valadez
- Department of Neurology, National Parkinson's Foundation Center for Excellence, Translational Neurotherapeutics Program. Movement Disorders Program. MedStar Georgetown Hospital Washington, DC, USA
| | - Yasar Torres-Yaghi
- Department of Neurology, National Parkinson's Foundation Center for Excellence, Translational Neurotherapeutics Program. Movement Disorders Program. MedStar Georgetown Hospital Washington, DC, USA
| | - Xu Huang
- Department of Neurology, Laboratory for Dementia and Parkinsonism, National Parkinson's Foundation Center for Excellence, Translational Neurotherapeutics Program. Georgetown University Medical Center, Washington, DC, USA
| | - Reversa R Mills
- Department of Neurology, National Parkinson's Foundation Center for Excellence, Translational Neurotherapeutics Program. Movement Disorders Program. MedStar Georgetown Hospital Washington, DC, USA
| | - Barbara M Wilmarth
- Department of Neurology, National Parkinson's Foundation Center for Excellence, Translational Neurotherapeutics Program. Movement Disorders Program. MedStar Georgetown Hospital Washington, DC, USA
| | - Hellen Howard
- Department of Neurology, National Parkinson's Foundation Center for Excellence, Translational Neurotherapeutics Program. Movement Disorders Program. MedStar Georgetown Hospital Washington, DC, USA
| | - Connell Dunn
- Department of Neurology, National Parkinson's Foundation Center for Excellence, Translational Neurotherapeutics Program. Movement Disorders Program. MedStar Georgetown Hospital Washington, DC, USA
| | - Alexis Carlson
- Department of Neurology, National Parkinson's Foundation Center for Excellence, Translational Neurotherapeutics Program. Movement Disorders Program. MedStar Georgetown Hospital Washington, DC, USA
| | - Abigail Lawler
- Department of Neurology, National Parkinson's Foundation Center for Excellence, Translational Neurotherapeutics Program. Movement Disorders Program. MedStar Georgetown Hospital Washington, DC, USA
| | - Sean L Rogers
- Department of Neurology, National Parkinson's Foundation Center for Excellence, Translational Neurotherapeutics Program. Movement Disorders Program. MedStar Georgetown Hospital Washington, DC, USA
| | - Ramsey A Falconer
- Department of Neurology, National Parkinson's Foundation Center for Excellence, Translational Neurotherapeutics Program. Movement Disorders Program. MedStar Georgetown Hospital Washington, DC, USA
| | - Jaeil Ahn
- Department of Biostatistics, Georgetown University Medical Center, Washington, DC, USA
| | - Zhaoxia Li
- Department of Neurology, National Parkinson's Foundation Center for Excellence, Translational Neurotherapeutics Program. Movement Disorders Program. MedStar Georgetown Hospital Washington, DC, USA
| | - Charbel Moussa
- Department of Neurology, Laboratory for Dementia and Parkinsonism, National Parkinson's Foundation Center for Excellence, Translational Neurotherapeutics Program. Georgetown University Medical Center, Washington, DC, USA
- Department of Neurology, National Parkinson's Foundation Center for Excellence, Translational Neurotherapeutics Program. Movement Disorders Program. MedStar Georgetown Hospital Washington, DC, USA
| |
Collapse
|
94
|
Garcia-Huerta P, Troncoso-Escudero P, Jerez C, Hetz C, Vidal RL. The intersection between growth factors, autophagy and ER stress: A new target to treat neurodegenerative diseases? Brain Res 2016; 1649:173-180. [PMID: 26993573 DOI: 10.1016/j.brainres.2016.02.052] [Citation(s) in RCA: 42] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2015] [Revised: 01/25/2016] [Accepted: 02/10/2016] [Indexed: 01/01/2023]
Abstract
One of the salient features of most neurodegenerative diseases is the aggregation of specific proteins in the brain. This proteostasis imbalance is proposed as a key event triggering the neurodegenerative cascade. The unfolded protein response (UPR) and autophagy pathways are emerging as critical processes implicated in handling disease-related misfolded proteins. However, in some conditions, perturbations in the buffering capacity of the proteostasis network may be part of the etiology of the disease. Thus, pharmacological or gene therapy strategies to enhance autophagy or UPR responses are becoming an attractive target for disease intervention. Here, we discuss current evidence depicting the complex involvement of autophagy and ER stress in brain diseases. Novel pathways to modulate protein misfolding are discussed including the relation between aging and growth factor signaling. This article is part of a Special Issue entitled SI:Autophagy.
Collapse
Affiliation(s)
- Paula Garcia-Huerta
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Program of Cellular and Molecular Biology, Center for Molecular Studies of the Cell Institute of Biomedical Sciences, University of Chile, Chile
| | - Paulina Troncoso-Escudero
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Program of Cellular and Molecular Biology, Center for Molecular Studies of the Cell Institute of Biomedical Sciences, University of Chile, Chile
| | - Carolina Jerez
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Neurounion Biomedical Foundation, Santiago, Chile
| | - Claudio Hetz
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Program of Cellular and Molecular Biology, Center for Molecular Studies of the Cell Institute of Biomedical Sciences, University of Chile, Chile; Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, USA.
| | - Rene L Vidal
- Biomedical Neuroscience Institute, Faculty of Medicine, University of Chile, Santiago, Chile; FONDAP Center for Geroscience, Brain Health and Metabolism, Santiago, Chile; Neurounion Biomedical Foundation, Santiago, Chile.
| |
Collapse
|
95
|
Edens BM, Miller N, Ma YC. Impaired Autophagy and Defective Mitochondrial Function: Converging Paths on the Road to Motor Neuron Degeneration. Front Cell Neurosci 2016; 10:44. [PMID: 26973461 PMCID: PMC4776126 DOI: 10.3389/fncel.2016.00044] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2015] [Accepted: 02/08/2016] [Indexed: 12/12/2022] Open
Abstract
Selective motor neuron degeneration is a hallmark of amyotrophic lateral sclerosis (ALS). Around 10% of all cases present as familial ALS (FALS), while sporadic ALS (SALS) accounts for the remaining 90%. Diverse genetic mutations leading to FALS have been identified, but the underlying causes of SALS remain largely unknown. Despite the heterogeneous and incompletely understood etiology, different types of ALS exhibit overlapping pathology and common phenotypes, including protein aggregation and mitochondrial deficiencies. Here, we review the current understanding of mechanisms leading to motor neuron degeneration in ALS as they pertain to disrupted cellular clearance pathways, ATP biogenesis, calcium buffering and mitochondrial dynamics. Through focusing on impaired autophagic and mitochondrial functions, we highlight how the convergence of diverse cellular processes and pathways contributes to common pathology in motor neuron degeneration.
Collapse
Affiliation(s)
- Brittany M. Edens
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of MedicineChicago, IL, USA
- Lurie Children’s Hospital of ChicagoChicago, IL, USA
| | - Nimrod Miller
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of MedicineChicago, IL, USA
- Lurie Children’s Hospital of ChicagoChicago, IL, USA
| | - Yong-Chao Ma
- Departments of Pediatrics, Neurology, and Physiology, Northwestern University Feinberg School of MedicineChicago, IL, USA
- Lurie Children’s Hospital of ChicagoChicago, IL, USA
| |
Collapse
|
96
|
Hebron M, Moussa CEH. Two sides of the same coin: tyrosine kinase inhibition in cancer and neurodegeneration. Neural Regen Res 2016; 10:1767-9. [PMID: 26807110 PMCID: PMC4705787 DOI: 10.4103/1673-5374.165320] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
Affiliation(s)
- Michaeline Hebron
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Georgetown University Medical Center, Washington D.C., NW, USA
| | - Charbel E-H Moussa
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Georgetown University Medical Center, Washington D.C., NW, USA
| |
Collapse
|
97
|
Zhang CW, Hang L, Yao TP, Lim KL. Parkin Regulation and Neurodegenerative Disorders. Front Aging Neurosci 2016; 7:248. [PMID: 26793099 PMCID: PMC4709595 DOI: 10.3389/fnagi.2015.00248] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2015] [Accepted: 12/17/2015] [Indexed: 12/13/2022] Open
Abstract
Parkin is a unique, multifunctional ubiquitin ligase whose various roles in the cell, particularly in neurons, are widely thought to be protective. The pivotal role that Parkin plays in maintaining neuronal survival is underscored by our current recognition that Parkin dysfunction represents not only a predominant cause of familial parkinsonism but also a formal risk factor for the more common, sporadic form of Parkinson’s disease (PD). Accordingly, keen research on Parkin over the past decade has led to an explosion of knowledge regarding its physiological roles and its relevance to PD. However, our understanding of Parkin is far from being complete. Indeed, surprises emerge from time to time that compel us to constantly update the paradigm of Parkin function. For example, we now know that Parkin’s function is not confined to mere housekeeping protein quality control (QC) roles but also includes mitochondrial homeostasis and stress-related signaling. Furthermore, emerging evidence also suggest a role for Parkin in several other major neurodegenerative diseases including Alzheimer’s disease (AD) and Amyotrophic Lateral Sclerosis (ALS). Yet, it remains truly amazing to note that a single enzyme could serve such multitude of functions and cellular roles. Clearly, its activity has to be tightly regulated. In this review, we shall discuss this and how dysregulated Parkin function may precipitate neuronal demise in various neurodegenerative disorders.
Collapse
Affiliation(s)
- Cheng-Wu Zhang
- Neurodegeneration Research Laboratory, National Neuroscience InstituteSingapore, Singapore; Institute of Advanced Materials, Nanjing Tech UniversityNanjing, People's Republic of China
| | - Liting Hang
- Department of Physiology, National University of Singapore Singapore, Singapore
| | - Tso-Pang Yao
- Departments of Pharmacology and Cancer Biology, Duke University Medical Center Durham, NC, USA
| | - Kah-Leong Lim
- Neurodegeneration Research Laboratory, National Neuroscience InstituteSingapore, Singapore; Institute of Advanced Materials, Nanjing Tech UniversityNanjing, People's Republic of China; Department of Physiology, National University of SingaporeSingapore, Singapore; Duke-NUS Graduate Medical School, National University of SingaporeSingapore, Singapore
| |
Collapse
|
98
|
Chauhan S, Ahmed Z, Bradfute SB, Arko-Mensah J, Mandell MA, Won Choi S, Kimura T, Blanchet F, Waller A, Mudd MH, Jiang S, Sklar L, Timmins GS, Maphis N, Bhaskar K, Piguet V, Deretic V. Pharmaceutical screen identifies novel target processes for activation of autophagy with a broad translational potential. Nat Commun 2015; 6:8620. [PMID: 26503418 PMCID: PMC4624223 DOI: 10.1038/ncomms9620] [Citation(s) in RCA: 116] [Impact Index Per Article: 12.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2015] [Accepted: 09/11/2015] [Indexed: 02/06/2023] Open
Abstract
Autophagy is a conserved homeostatic process active in all human cells and affecting a spectrum of diseases. Here we use a pharmaceutical screen to discover new mechanisms for activation of autophagy. We identify a subset of pharmaceuticals inducing autophagic flux with effects in diverse cellular systems modelling specific stages of several human diseases such as HIV transmission and hyperphosphorylated tau accumulation in Alzheimer's disease. One drug, flubendazole, is a potent inducer of autophagy initiation and flux by affecting acetylated and dynamic microtubules in a reciprocal way. Disruption of dynamic microtubules by flubendazole results in mTOR deactivation and dissociation from lysosomes leading to TFEB (transcription factor EB) nuclear translocation and activation of autophagy. By inducing microtubule acetylation, flubendazole activates JNK1 leading to Bcl-2 phosphorylation, causing release of Beclin1 from Bcl-2-Beclin1 complexes for autophagy induction, thus uncovering a new approach to inducing autophagic flux that may be applicable in disease treatment. Autophagy is a homeostatic process that could be a potential drug target in the treatment of disease. Here the authors identify in a pharmaceutical screen flubendazole as an inducer of autophagy initiation and flux by affecting microtubules, mTOR, TFEB and Beclin 1 activity.
Collapse
Affiliation(s)
- Santosh Chauhan
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Zahra Ahmed
- Cardiff Institute of Infection &Immunity, Cardiff University, School of Medicine, Henry Wellcome Building, Heath Park CF14 4XN, Cardiff, UK
| | - Steven B Bradfute
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - John Arko-Mensah
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Michael A Mandell
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Seong Won Choi
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Tomonori Kimura
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Fabien Blanchet
- Cardiff Institute of Infection &Immunity, Cardiff University, School of Medicine, Henry Wellcome Building, Heath Park CF14 4XN, Cardiff, UK
| | - Anna Waller
- Department of Pathology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Michal H Mudd
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Shanya Jiang
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Larry Sklar
- Department of Pathology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Graham S Timmins
- College of Pharmacy, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Nicole Maphis
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Kiran Bhaskar
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA.,Department of Neurology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| | - Vincent Piguet
- Cardiff Institute of Infection &Immunity, Cardiff University, School of Medicine, Henry Wellcome Building, Heath Park CF14 4XN, Cardiff, UK
| | - Vojo Deretic
- Department of Molecular Genetics and Microbiology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA.,Department of Neurology, School of Medicine, University of New Mexico Health Sciences Center, 915 Camino de Salud, NE, Albuquerque, New Mexico 87131, USA
| |
Collapse
|
99
|
Passeri E, Mocchetti I, Moussa C. Is human immunodeficiency virus-mediated dementia an autophagic defect that leads to neurodegeneration? CNS & NEUROLOGICAL DISORDERS-DRUG TARGETS 2015; 13:1571-9. [PMID: 25106633 DOI: 10.2174/1871527313666140806125841] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/05/2013] [Revised: 04/04/2014] [Accepted: 06/06/2014] [Indexed: 11/22/2022]
Abstract
Autophagy is a cellular process that mediates selective degradation of cellular components in lysosomes. Autophagy may protect against neuronal apoptosis, which is induced in a number of neurodegenerative diseases. Thus, compounds that modulate autophagy could be beneficial to treat neurological disorders characterized by apoptosis such as Parkinson's and Alzheimer's diseases, as well as human-immunodeficiency virus-dementia complex. In this paper, we review new and old evidence on the role of autophagy in neuronal cell survival and we present evidence that humanimmunodeficiency virus may have adapted strategies to alter autophagic pathways in neurons. Moreover, we discuss the usefulness of drugs that facilitate autophagic clearance of proteins that are associated with neurodegeneration.
Collapse
Affiliation(s)
| | | | - Charbel Moussa
- Georgetown University Medical Center, Department of Neuroscience, NRB WP13, 3970 Reservoir Rd, NW, Washington, DC 20057, USA.
| |
Collapse
|
100
|
Lonskaya I, Hebron ML, Selby ST, Turner RS, Moussa CEH. Nilotinib and bosutinib modulate pre-plaque alterations of blood immune markers and neuro-inflammation in Alzheimer's disease models. Neuroscience 2015; 304:316-27. [PMID: 26235435 DOI: 10.1016/j.neuroscience.2015.07.070] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2015] [Revised: 07/15/2015] [Accepted: 07/25/2015] [Indexed: 01/08/2023]
Abstract
Alzheimer's disease (AD) brains exhibit plaques and tangles in association with inflammation. The non-receptor tyrosine kinase Abl is linked to neuro-inflammation in AD. Abl inhibition by nilotinib or bosutinib facilitates amyloid clearance and may decrease inflammation. Transgenic mice that express Dutch, Iowa and Swedish APP mutations (TgAPP) and display progressive Aβ plaque deposition were treated with tyrosine kinase inhibitors (TKIs) to determine pre-plaque effects on systemic and CNS inflammation using milliplex® ELISA. Plaque Aβ was detected at 4months in TgAPP and pre-plaque intracellular Aβ accumulation (2.5months) was associated with changes of cytokines and chemokines prior to detection of glial changes. Plaque formation correlated with increased levels of pro-inflammatory cytokines (TNF-α, IL-6, IL-1α, IL-1β) and markers of immunosuppressive and adaptive immunity, including, IL-4, IL-10, IL-2, IL-3, Vascular Endothelial Growth Factor (VEGF) and IFN-γ. An inverse relationship of chemokines was observed as CCL2 and CCL5 were lower than WT mice at 2months and significantly increased after plaque appearance, while soluble CX3CL1 decreased. A change in glial profile was only robustly detected at 6months in Tg-APP mice and TKIs reduced astrocyte and dendritic cell number with no effects on microglia, suggesting alteration of brain immunity. Nilotinib decreased blood and brain cytokines and chemokines and increased CX3CL1. Bosutinib increased brain and blood IL-10 and CX3CL1, suggesting a protective role for soluble CX3CL1. Taken together these data suggest that TKIs regulate systemic and CNS immunity and may be useful treatments in early AD through dual effects on amyloid clearance and immune modulation.
Collapse
Affiliation(s)
- I Lonskaya
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Georgetown University Medical Center, Washington D.C. 20007, USA
| | - M L Hebron
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Georgetown University Medical Center, Washington D.C. 20007, USA
| | - S T Selby
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Georgetown University Medical Center, Washington D.C. 20007, USA
| | - R S Turner
- Department of Neurology, Memory Disorders Program, Georgetown University Medical Center, Washington D.C. 20007, USA
| | - C E-H Moussa
- Department of Neurology, Laboratory for Dementia and Parkinsonism, Georgetown University Medical Center, Washington D.C. 20007, USA.
| |
Collapse
|