101
|
Modulation of proteostasis and protein trafficking: a therapeutic avenue for misfolded G protein-coupled receptors causing disease in humans. Emerg Top Life Sci 2019; 3:39-52. [PMID: 33523195 DOI: 10.1042/etls20180055] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2018] [Revised: 02/20/2019] [Accepted: 02/21/2019] [Indexed: 11/17/2022]
Abstract
Proteostasis refers to the process whereby the cell maintains in equilibrium the protein content of different compartments. This system consists of a highly interconnected network intended to efficiently regulate the synthesis, folding, trafficking, and degradation of newly synthesized proteins. Molecular chaperones are key players of the proteostasis network. These proteins assist in the assembly and folding processes of newly synthesized proteins in a concerted manner to achieve a three-dimensional structure compatible with export from the endoplasmic reticulum to other cell compartments. Pharmacologic interventions intended to modulate the proteostasis network and tackle the devastating effects of conformational diseases caused by protein misfolding are under development. These include small molecules called pharmacoperones, which are highly specific toward the target protein serving as a molecular framework to cause misfolded mutant proteins to fold and adopt a stable conformation suitable for passing the scrutiny of the quality control system and reach its correct location within the cell. Here, we review the main components of the proteostasis network and how pharmacoperones may be employed to correct misfolding of two G protein-coupled receptors, the vasopressin 2 receptor and the gonadotropin-releasing hormone receptor, whose mutations lead to X-linked nephrogenic diabetes insipidus and congenital hypogonadotropic hypogonadism in humans respectively.
Collapse
|
102
|
Wang Y, Kim B, Walker A, Williams S, Meeks A, Lee YJ, Seo SS. Cytotoxic effects of parathion, paraoxon, and their methylated derivatives on a mouse neuroblastoma cell line NB41A3. ACTA ACUST UNITED AC 2019. [DOI: 10.2131/fts.6.45] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Affiliation(s)
- Yunbiao Wang
- Department of Chemistry and Forensic Science, Albany State University, USA
| | - ByungHoon Kim
- Department of Biological Sciences, Albany State University, USA
| | - Ashley Walker
- Department of Chemistry and Forensic Science, Albany State University, USA
| | - Shayla Williams
- Department of Biological Sciences, Albany State University, USA
| | - Ashley Meeks
- Department of Chemistry and Forensic Science, Albany State University, USA
| | - Yong-Jin Lee
- Department of Biological Sciences, Albany State University, USA
| | - Seong S. Seo
- Department of Chemistry and Forensic Science, Albany State University, USA
| |
Collapse
|
103
|
Moura J, Madureira P, Leal EC, Fonseca AC, Carvalho E. Immune aging in diabetes and its implications in wound healing. Clin Immunol 2019; 200:43-54. [PMID: 30735729 PMCID: PMC7322932 DOI: 10.1016/j.clim.2019.02.002] [Citation(s) in RCA: 63] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2019] [Revised: 02/04/2019] [Accepted: 02/04/2019] [Indexed: 02/06/2023]
Abstract
Immune systems have evolved to recognize and eliminate pathogens and damaged cells. In humans, it is estimated to recognize 109 epitopes and natural selection ensures that clonally expanded cells replace unstimulated cells and overall immune cell numbers remain stationary. But, with age, it faces continuous repertoire restriction and concomitant accumulation of primed cells. Changes shaping the aging immune system have bitter consequences because, as inflammatory responses gain intensity and duration, tissue-damaging immunity and inflammatory disease arise. During inflammation, the glycolytic flux cannot cope with increasing ATP demands, limiting the immune response's extent. In diabetes, higher glucose availability stretches the glycolytic limit, dysregulating proteostasis and increasing T-cell expansion. Long-term hyperglycemia exerts an accumulating effect, leading to higher inflammatory cytokine levels and increased cytotoxic mediator secretion upon infection, a phenomenon known as diabetic chronic inflammation. Here we review the etiology of diabetic chronic inflammation and its consequences on wound healing.
Collapse
Affiliation(s)
- J Moura
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; INEB - Instituto Nacional de Engenharia Biomédica, University of Porto, Porto, Portugal; i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal.
| | - P Madureira
- i3S - Instituto de Investigação e Inovação em Saúde, University of Porto, Porto, Portugal; IBMC - Instituto de Biologia Celular e Molecular, University of Porto, Porto, Portugal; Immunethep, Biocant Park, Cantanhede, Portugal
| | - E C Leal
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - A C Fonseca
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal
| | - E Carvalho
- Center for Neuroscience and Cell Biology, University of Coimbra, Coimbra, Portugal; Instituto de Investigação Interdisciplinar, University of Coimbra, Coimbra, Portugal; Department of Geriatrics, University of Arkansas for Medical Sciences and Arkansas Children's Research Institute, Little Rock, AR, United States
| |
Collapse
|
104
|
Soares TR, Reis SD, Pinho BR, Duchen MR, Oliveira JMA. Targeting the proteostasis network in Huntington's disease. Ageing Res Rev 2019; 49:92-103. [PMID: 30502498 PMCID: PMC6320389 DOI: 10.1016/j.arr.2018.11.006] [Citation(s) in RCA: 49] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2018] [Revised: 11/19/2018] [Accepted: 11/26/2018] [Indexed: 12/31/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant neurodegenerative disorder caused by a polyglutamine expansion mutation in the huntingtin protein. Expansions above 40 polyglutamine repeats are invariably fatal, following a symptomatic period characterised by choreiform movements, behavioural abnormalities, and cognitive decline. While mutant huntingtin (mHtt) is widely expressed from early life, most patients with HD present in mid-adulthood, highlighting the role of ageing in disease pathogenesis. mHtt undergoes proteolytic cleavage, misfolding, accumulation, and aggregation into inclusion bodies. The emerging model of HD pathogenesis proposes that the chronic production of misfolded mHtt overwhelms the chaperone machinery, diverting other misfolded clients to the proteasome and the autophagy pathways, ultimately leading to a global collapse of the proteostasis network. Multiple converging hypotheses also implicate ageing and its impact in the dysfunction of organelles as additional contributing factors to the collapse of proteostasis in HD. In particular, mitochondrial function is required to sustain the activity of ATP-dependent chaperones and proteolytic machinery. Recent studies elucidating mitochondria-endoplasmic reticulum interactions and uncovering a dedicated proteostasis machinery in mitochondria, suggest that mitochondria play a more active role in the maintenance of cellular proteostasis than previously thought. The enhancement of cytosolic proteostasis pathways shows promise for HD treatment, protecting cells from the detrimental effects of mHtt accumulation. In this review, we consider how mHtt and its post translational modifications interfere with protein quality control pathways, and how the pharmacological and genetic modulation of components of the proteostasis network impact disease phenotypes in cellular and in vivo HD models.
Collapse
Affiliation(s)
- Tânia R Soares
- REQUIMTE/LAQV, Department of Drug Sciences, Pharmacology Lab, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal; Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK
| | - Sara D Reis
- REQUIMTE/LAQV, Department of Drug Sciences, Pharmacology Lab, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Brígida R Pinho
- REQUIMTE/LAQV, Department of Drug Sciences, Pharmacology Lab, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal
| | - Michael R Duchen
- Department of Cell and Developmental Biology, University College London, London, WC1E 6BT, UK; Consortium for Mitochondrial Research (CfMR), University College London, Gower Street, WC1E 6BT, London, UK
| | - Jorge M A Oliveira
- REQUIMTE/LAQV, Department of Drug Sciences, Pharmacology Lab, Faculty of Pharmacy, University of Porto, 4050-313, Porto, Portugal; Consortium for Mitochondrial Research (CfMR), University College London, Gower Street, WC1E 6BT, London, UK.
| |
Collapse
|
105
|
Linking unfolded protein response to inflammation and depression: potential pathologic and therapeutic implications. Mol Psychiatry 2019; 24:987-994. [PMID: 30214045 PMCID: PMC6416085 DOI: 10.1038/s41380-018-0241-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/24/2018] [Revised: 06/26/2018] [Accepted: 08/16/2018] [Indexed: 11/21/2022]
Abstract
Depression is a devastating mental disorder that affects millions of people worldwide. Inflammation has been shown to be a key factor involved in the underlying pathophysiology of depression and has been shown in a substantial proportion of cases of depression. Changes attributed with morphological deformities and immunomodulation in susceptible regions of the depressed brain raised the possibility of altered cellular homeostasis transduced by the intracellular stress response. How emotional stressors can lead to an inflamed brain that directly affects physiology and activity is yet to be fully understood. The unfolded protein response (UPR) has been shown to be active in both models of depression as well as in postmortem brain of depressed individuals. The UPR is the cellular response to stress which results in misfolded proteins. Interestingly, UPR activation is directly linked to both inflammatory cytokine production and Toll-like receptor (TLR) expression. The TLRs are part of the innate immune response which typically reacts to "classic invasions" such as bacteria or viruses as well as trauma. TLRs have also been shown to be upregulated in depression, thus solidifying the connection between inflammation and depression. In this review, we aim to tie the UPR-TLR response and depression, and describe the implications of such an association. We also propose future directions for their role in treatment for depression.
Collapse
|
106
|
Long JM, Maloney B, Rogers JT, Lahiri DK. Novel upregulation of amyloid-β precursor protein (APP) by microRNA-346 via targeting of APP mRNA 5'-untranslated region: Implications in Alzheimer's disease. Mol Psychiatry 2019; 24:345-363. [PMID: 30470799 PMCID: PMC6514885 DOI: 10.1038/s41380-018-0266-3] [Citation(s) in RCA: 114] [Impact Index Per Article: 19.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/18/2018] [Revised: 07/27/2018] [Accepted: 09/06/2018] [Indexed: 12/14/2022]
Abstract
In addition to the devastating symptoms of dementia, Alzheimer's disease (AD) is characterized by accumulation of the processing products of the amyloid-β (Aβ) peptide precursor protein (APP). APP's non-pathogenic functions include regulating intracellular iron (Fe) homeostasis. MicroRNAs are small (~ 20 nucleotides) RNA species that instill specificity to the RNA-induced silencing complex (RISC). In most cases, RISC inhibits mRNA translation through the 3'-untranslated region (UTR) sequence. By contrast, we report a novel activity of miR-346: specifically, that it targets the APP mRNA 5'-UTR to upregulate APP translation and Aβ production. This upregulation is reduced but not eliminated by knockdown of argonaute 2. The target site for miR-346 overlaps with active sites for an iron-responsive element (IRE) and an interleukin-1 (IL-1) acute box element. IREs interact with iron response protein1 (IRP1), an iron-dependent translational repressor. In primary human brain cultures, miR-346 activity required chelation of Fe. In addition, miR-346 levels are altered in late-Braak stage AD. Thus, miR-346 plays a role in upregulation of APP in the CNS and participates in maintaining APP regulation of Fe, which is disrupted in late stages of AD. Further work will be necessary to integrate other metals, and IL-1 into the Fe-miR-346 activity network. We, thus, propose a "FeAR" (Fe, APP, RNA) nexus in the APP 5'-UTR that includes an overlapping miR-346-binding site and the APP IRE. When a "healthy FeAR" exists, activities of miR-346 and IRP/Fe interact to maintain APP homeostasis. Disruption of an element that targets the FeAR nexus would lead to pathogenic disruption of APP translation and protein production.
Collapse
Affiliation(s)
- Justin M. Long
- 0000 0001 2287 3919grid.257413.6Department of Psychiatry, Laboratory of Molecular Neurogenetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Bryan Maloney
- 0000 0001 2287 3919grid.257413.6Department of Psychiatry, Laboratory of Molecular Neurogenetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA ,0000 0001 2287 3919grid.257413.6Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| | - Jack T. Rogers
- Neurochemistry Laboratory, Department of Psychiatry-Neuroscience, MGH, Harvard Medical School, Charlestown, MA 02129 USA
| | - Debomoy K. Lahiri
- 0000 0001 2287 3919grid.257413.6Department of Psychiatry, Laboratory of Molecular Neurogenetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA ,0000 0001 2287 3919grid.257413.6Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, IN 46202 USA ,0000 0001 2287 3919grid.257413.6Department of Medical and Molecular Genetics, Indiana University School of Medicine, Indianapolis, IN 46202 USA ,0000 0001 2287 3919grid.257413.6Indiana Alzheimer Disease Center, Indiana University School of Medicine, Indianapolis, IN 46202 USA
| |
Collapse
|
107
|
Yamaguchi Y, Oh-hashi K, Matsuoka Y, Takemura H, Yamakita S, Matsuda M, Sawa T, Amaya F. Endoplasmic Reticulum Stress in the Dorsal Root Ganglion Contributes to the Development of Pain Hypersensitivity after Nerve Injury. Neuroscience 2018; 394:288-299. [DOI: 10.1016/j.neuroscience.2018.08.005] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/07/2017] [Revised: 07/05/2018] [Accepted: 08/07/2018] [Indexed: 12/30/2022]
|
108
|
Agarwal N. Impact of Endoplasmic Reticulum Stress in Development of Pain Hypersensitivity: A Commentary on Yamaguchi et al. Neuroscience 2018; 394:286-287. [PMID: 30366026 DOI: 10.1016/j.neuroscience.2018.10.034] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Accepted: 10/15/2018] [Indexed: 10/28/2022]
Affiliation(s)
- Nitin Agarwal
- Pharmacology Institute, Heidelberg University, Im Neuenheimer Feld 366, 69120 Heidelberg, Germany.
| |
Collapse
|
109
|
Abstract
The brain undergoes several changes at structural, molecular, and cellular levels leading to alteration in its functions and these processes are primarily maintained by proteostasis in cells. However, an imbalance in proteostasis due to the abnormal accumulation of protein aggregates induces endoplasmic reticulum (ER) stress. This event, in turn, activate the unfolded protein response; however, in most neurodegenerative conditions and brain injury, an uncontrolled unfolded protein response elicits memory dysfunction. Although the underlying signaling mechanism for impairment of memory function following induction of ER stress remains elusive, recent studies have highlighted that inactivation of a transcription factor, CREB, which is essential for synaptic function and memory formation, plays an essential role for ER stress-induced synaptic and memory dysfunction. In this review, current studies and most updated view on how ER stress affects memory function in both physiological and pathological conditions will be highlighted.
Collapse
Affiliation(s)
- Nilkantha Sen
- Department of Neurological Surgery, University of Pittsburgh, Pittsburgh, PA, USA
| |
Collapse
|
110
|
García-González P, Cabral-Miranda F, Hetz C, Osorio F. Interplay Between the Unfolded Protein Response and Immune Function in the Development of Neurodegenerative Diseases. Front Immunol 2018; 9:2541. [PMID: 30450103 PMCID: PMC6224445 DOI: 10.3389/fimmu.2018.02541] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 10/15/2018] [Indexed: 12/25/2022] Open
Abstract
Emerging evidence suggests that the immune and nervous systems are in close interaction in health and disease conditions. Protein aggregation and proteostasis dysfunction at the level of the endoplasmic reticulum (ER) are central contributors to neurodegenerative diseases. The unfolded protein response (UPR) is the main transduction pathway that maintains protein homeostasis under conditions of protein misfolding and aggregation. Brain inflammation often coexists with the degenerative process in different brain diseases. Interestingly, besides its well-described role in neuronal fitness, the UPR has also emerged as a key regulator of ontogeny and function of several immune cell types. Nevertheless, the contribution of the UPR to brain inflammation initiated by immune cells remains largely unexplored. In this review, we provide a perspective on the potential role of ER stress signaling in brain-associated immune cells and the possible implications to neuroinflammation and development of neurodegenerative diseases.
Collapse
Affiliation(s)
- Paulina García-González
- Laboratory of Immunology and Cellular Stress, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| | - Felipe Cabral-Miranda
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile.,Brain Health and Metabolism, FONDAP Center for Geroscience, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Instituto de Ciências Biomédicas, Universidade Federal do Rio de Janeiro, Rio de Janeiro, Brazil
| | - Claudio Hetz
- Faculty of Medicine, Biomedical Neuroscience Institute, University of Chile, Santiago, Chile.,Brain Health and Metabolism, FONDAP Center for Geroscience, Santiago, Chile.,Program of Cellular and Molecular Biology, Institute of Biomedical Sciences, University of Chile, Santiago, Chile.,Buck Institute for Research on Aging, Novato, CA, United States.,Department of Immunology and Infectious Diseases, Harvard School of Public Health, Boston, MA, United States
| | - Fabiola Osorio
- Laboratory of Immunology and Cellular Stress, Program of Immunology, Institute of Biomedical Sciences, Faculty of Medicine, University of Chile, Santiago, Chile
| |
Collapse
|
111
|
Cheng J, North BJ, Zhang T, Dai X, Tao K, Guo J, Wei W. The emerging roles of protein homeostasis-governing pathways in Alzheimer's disease. Aging Cell 2018; 17:e12801. [PMID: 29992725 PMCID: PMC6156496 DOI: 10.1111/acel.12801] [Citation(s) in RCA: 75] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/29/2018] [Accepted: 06/04/2018] [Indexed: 12/22/2022] Open
Abstract
Pathways governing protein homeostasis are involved in maintaining the structural, quantitative, and functional stability of intracellular proteins and involve the ubiquitin-proteasome system, autophagy, endoplasmic reticulum, and mTOR pathway. Due to the broad physiological implications of protein homeostasis pathways, dysregulation of proteostasis is often involved in the development of multiple pathological conditions, including Alzheimer's disease (AD). Similar to other neurodegenerative diseases that feature pathogenic accumulation of misfolded proteins, Alzheimer's disease is characterized by two pathological hallmarks, amyloid-β (Aβ) plaques and tau aggregates. Knockout or transgenic overexpression of various proteostatic components in mice results in AD-like phenotypes. While both Aβ plaques and tau aggregates could in turn enhance the dysfunction of these proteostatic pathways, eventually leading to apoptotic or necrotic neuronal death and pathogenesis of Alzheimer's disease. Therefore, targeting the components of proteostasis pathways may be a promising therapeutic strategy against Alzheimer's disease.
Collapse
Affiliation(s)
- Ji Cheng
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
- Department of PathologyBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusetts
| | - Brian J. North
- Department of PathologyBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusetts
| | - Tao Zhang
- Department of PathologyBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusetts
| | - Xiangpeng Dai
- Department of PathologyBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusetts
| | - Kaixiong Tao
- Department of Gastrointestinal SurgeryUnion HospitalTongji Medical CollegeHuazhong University of Science and TechnologyWuhanChina
| | - Jianping Guo
- Department of PathologyBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusetts
| | - Wenyi Wei
- Department of PathologyBeth Israel Deaconess Medical CenterHarvard Medical SchoolBostonMassachusetts
| |
Collapse
|
112
|
Charvin D, Medori R, Hauser RA, Rascol O. Therapeutic strategies for Parkinson disease: beyond dopaminergic drugs. Nat Rev Drug Discov 2018; 17:804-822. [PMID: 30262889 DOI: 10.1038/nrd.2018.136] [Citation(s) in RCA: 165] [Impact Index Per Article: 23.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Existing therapeutic strategies for managing Parkinson disease (PD), which focus on addressing the loss of dopamine and dopaminergic function linked with degeneration of dopaminergic neurons, are limited by side effects and lack of long-term efficacy. In recent decades, research into PD pathophysiology and pharmacology has focused on understanding and tackling the neurodegenerative processes and symptomology of PD. In this Review, we discuss the challenges associated with the development of novel therapies for PD, highlighting emerging agents that aim to target cell death, as well as new targets offering a symptomatic approach to managing features and progression of the disease.
Collapse
Affiliation(s)
| | | | - Robert A Hauser
- Department of Neurology, University of South Florida, Tampa, FL, USA
| | - Olivier Rascol
- Centre d'Investigation Clinique CIC1436, Services de Neurologie et de Pharmacologie Clinique, Réseau NS-PARK/FCRIN et Centre COEN NeuroToul, CHU de Toulouse, INSERM, University of Toulouse 3, Toulouse, France
| |
Collapse
|
113
|
Selvakumar GP, Iyer SS, Kempuraj D, Ahmed ME, Thangavel R, Dubova I, Raikwar SP, Zaheer S, Zaheer A. Molecular Association of Glia Maturation Factor with the Autophagic Machinery in Rat Dopaminergic Neurons: a Role for Endoplasmic Reticulum Stress and MAPK Activation. Mol Neurobiol 2018; 56:3865-3881. [PMID: 30218400 DOI: 10.1007/s12035-018-1340-1] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2018] [Accepted: 08/30/2018] [Indexed: 12/16/2022]
Abstract
Parkinson's disease (PD) is one of the several neurodegenerative diseases where accumulation of aggregated proteins like α-synuclein occurs. Dysfunction in autophagy leading to this protein build-up and subsequent dopaminergic neurodegeneration may be one of the causes of PD. The mechanisms that impair autophagy remain poorly understood. 1-Methyl-4-phenylpiridium ion (MPP+) is a neurotoxin that induces experimental PD in vitro. Our studies have shown that glia maturation factor (GMF), a brain-localized inflammatory protein, induces dopaminergic neurodegeneration in PD and that suppression of GMF prevents MPP+-induced loss of dopaminergic neurons. In the present study, we demonstrate a molecular action of GMF on the autophagic machinery resulting in dopaminergic neuronal loss and propose GMF-mediated autophagic dysfunction as one of the contributing factors in PD progression. Using dopaminergic N27 neurons, primary neurons from wild type (WT), and GMF-deficient (GMF-KO) mice, we show that GMF and MPP+ enhanced expression of MAPKs increased the mammalian target of rapamycin (mTOR) activation and endoplasmic reticulum stress markers such as phospho-eukaryotic translation initiation factor 2 alpha kinase 3 (p-PERK) and inositol-requiring enzyme 1α (IRE1α). Further, GMF and MPP+ reduced Beclin 1, focal adhesion kinase (FAK) family-interacting protein of 200 kD (FIP200), and autophagy-related proteins (ATGs) 3, 5, 7, 16L, and 12. The combined results demonstrate that GMF affects autophagy through autophagosome formation with significantly reduced lysosomal-associated membrane protein 1/2, and the number of autophagic acidic vesicles. Using primary neurons, we show that MPP+ treatment leads to differential expression and localization of p62/sequestosome and in GMF-KO neurons, there was a marked increase in p62 staining implying autophagy deficiency with very little co-localization of α-synuclein and p62 as compared with WT neurons. Collectively, this study provides a bidirectional role for GMF in executing dopaminergic neuronal death mediated by autophagy that is relevant to PD.
Collapse
Affiliation(s)
- Govindhasamy Pushpavathi Selvakumar
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.,Department of Neurology, and Center for Translational Neuroscience, School of Medicine-University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, USA
| | - Shankar S Iyer
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.,Department of Neurology, and Center for Translational Neuroscience, School of Medicine-University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, USA
| | - Duraisamy Kempuraj
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.,Department of Neurology, and Center for Translational Neuroscience, School of Medicine-University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, USA
| | - Mohammad Ejaz Ahmed
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.,Department of Neurology, and Center for Translational Neuroscience, School of Medicine-University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, USA
| | - Ramasamy Thangavel
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.,Department of Neurology, and Center for Translational Neuroscience, School of Medicine-University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, USA
| | - Iuliia Dubova
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Sudhanshu P Raikwar
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA.,Department of Neurology, and Center for Translational Neuroscience, School of Medicine-University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, USA
| | - Smita Zaheer
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA
| | - Asgar Zaheer
- Harry S. Truman Memorial Veterans Hospital, Columbia, MO, USA. .,Department of Neurology, and Center for Translational Neuroscience, School of Medicine-University of Missouri, M741A Medical Science Building, 1 Hospital Drive, Columbia, MO, USA.
| |
Collapse
|
114
|
Mechanism of aggregation and membrane interactions of mammalian prion protein. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018. [DOI: 10.1016/j.bbamem.2018.02.031] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
|
115
|
Chiu CC, Lu CS, Weng YH, Chen YL, Huang YZ, Chen RS, Cheng YC, Huang YC, Liu YC, Lai SC, Lin KJ, Lin YW, Chen YJ, Chen CL, Yeh TH, Wang HL. PARK14 (D331Y) PLA2G6 Causes Early-Onset Degeneration of Substantia Nigra Dopaminergic Neurons by Inducing Mitochondrial Dysfunction, ER Stress, Mitophagy Impairment and Transcriptional Dysregulation in a Knockin Mouse Model. Mol Neurobiol 2018; 56:3835-3853. [PMID: 30088174 DOI: 10.1007/s12035-018-1118-5] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2018] [Accepted: 05/11/2018] [Indexed: 12/27/2022]
Abstract
PARK14 patients with homozygous (D331Y) PLA2G6 mutation display motor deficits of pure early-onset Parkinson's disease (PD). The aim of this study is to investigate the pathogenic mechanism of mutant (D331Y) PLA2G6-induced PD. We generated knockin (KI) mouse model of PARK14 harboring homozygous (D331Y) PLA2G6 mutation. Then, we investigated neuropathological and neurological phenotypes of PLA2G6D331Y/D331Y KI mice and molecular pathogenic mechanisms of (D331Y) PLA2G6-induced degeneration of substantia nigra (SN) dopaminergic neurons. Six-or nine-month-old PLA2G6D331Y/D331Y KI mice displayed early-onset cell death of SNpc dopaminergic neurons. Lewy body pathology was found in the SN of PLA2G6D331Y/D331Y mice. Six-or nine-month-old PLA2G6D331Y/D331Y KI mice exhibited early-onset parkinsonism phenotypes. Disrupted cristae of mitochondria were found in SNpc dopaminergic neurons of PLA2G6D331Y/D331Y mice. PLA2G6D331Y/D331Y mice displayed mitochondrial dysfunction and upregulated ROS production, which may lead to activation of apoptotic cascade. Upregulated protein levels of Grp78, IRE1, PERK, and CHOP, which are involved in activation of ER stress, were found in the SN of PLA2G6D331Y/D331Y mice. Protein expression of mitophagic proteins, including parkin and BNIP3, was downregulated in the SN of PLA2G6D331Y/D331Y mice, suggesting that (D331Y) PLA2G6 mutation causes mitophagy dysfunction. In the SN of PLA2G6D331Y/D331Y mice, mRNA levels of eight genes that are involved in neuroprotection/neurogenesis were decreased, while mRNA levels of two genes that promote apoptotic death were increased. Our results suggest that PARK14 (D331Y) PLA2G6 mutation causes degeneration of SNpc dopaminergic neurons by causing mitochondrial dysfunction, elevated ER stress, mitophagy impairment, and transcriptional abnormality.
Collapse
Affiliation(s)
- Ching-Chi Chiu
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan.,Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Chin-Song Lu
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University College of Medicine, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Hsin Weng
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University College of Medicine, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Ying-Ling Chen
- Department of Nursing, Chang Gung University of Science and Technology, Taoyuan, Taiwan
| | - Ying-Zu Huang
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University College of Medicine, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Institute of Cognitive Neuroscience, National Central University, Taoyuan, Taiwan
| | - Rou-Shayn Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University College of Medicine, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Yi-Chuan Cheng
- Graduate Institute of Biomedical Sciences, Chang Gung University College of Medicine, Taoyuan, Taiwan
| | - Yin-Cheng Huang
- College of Medicine, Chang Gung University, Taoyuan, Taiwan.,Department of Neurosurgery, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yu-Chuan Liu
- Division of Sports Medicine, Taiwan Landseed Hospital, Taoyuan, Taiwan
| | - Szu-Chia Lai
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan.,Healthy Aging Research Center, Chang Gung University College of Medicine, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan.,College of Medicine, Chang Gung University, Taoyuan, Taiwan
| | - Kun-Jun Lin
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan.,Molecular Imaging Center, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yan-Wei Lin
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan.,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan
| | - Yu-Jie Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan
| | - Chao-Lang Chen
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan
| | - Tu-Hsueh Yeh
- Department of Neurology, Taipei Medical University Hospital, No. 252, Wuxing St, Xinyi District, Taipei City, 110, Taiwan. .,School of Medicine, Taipei Medical University, Taipei, Taiwan.
| | - Hung-Li Wang
- Neuroscience Research Center, Chang Gung Memorial Hospital at Linkou, Linkou, Taoyuan, Taiwan. .,Healthy Aging Research Center, Chang Gung University College of Medicine, Taoyuan, Taiwan. .,Division of Movement Disorders, Department of Neurology, Chang Gung Memorial Hospital at Linkou, Taoyuan, Taiwan. .,Department of Physiology and Pharmacology, Chang Gung University College of Medicine, No. 259, Wen-Hwa 1st Road, Kweishan, Taoyuan, 333, Taiwan.
| |
Collapse
|
116
|
Yao J, Qiu Y, Frontera E, Jia L, Khan NW, Klionsky DJ, Ferguson TA, Thompson DA, Zacks DN. Inhibiting autophagy reduces retinal degeneration caused by protein misfolding. Autophagy 2018; 14:1226-1238. [PMID: 29940785 PMCID: PMC6103695 DOI: 10.1080/15548627.2018.1463121] [Citation(s) in RCA: 67] [Impact Index Per Article: 9.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2017] [Revised: 04/02/2018] [Accepted: 04/04/2018] [Indexed: 12/13/2022] Open
Abstract
Mutations in the genes necessary for the structure and function of vertebrate photoreceptor cells are associated with multiple forms of inherited retinal degeneration. Mutations in the gene encoding RHO (rhodopsin) are a common cause of autosomal dominant retinitis pigmentosa (adRP), with the Pro23His variant of RHO resulting in a misfolded protein that activates endoplasmic reticulum stress and the unfolded protein response. Stimulating macroautophagy/autophagy has been proposed as a strategy for clearing misfolded RHO and reducing photoreceptor death. We found that retinas from mice heterozygous for the gene encoding the RHOP23H variant (hereafter called P23H) exhibited elevated levels of autophagy flux, and that pharmacological stimulation of autophagy accelerated retinal degeneration. In contrast, reducing autophagy flux pharmacologically or by rod-specific deletion of the autophagy-activating gene Atg5, improved photoreceptor structure and function. Furthermore, proteasome levels and activity were reduced in the P23H retina, and increased when Atg5 was deleted. Our findings suggest that autophagy contributes to photoreceptor cell death in P23H mice, and that decreasing autophagy shifts the degradation of misfolded RHO protein to the proteasome and is protective. These observations suggest that modulating the flux of misfolded proteins from autophagy to the proteasome may represent an important therapeutic strategy for reducing proteotoxicity in adRP and other diseases caused by protein folding defects.
Collapse
Affiliation(s)
- Jingyu Yao
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Yaoyan Qiu
- Department of Ophthalmology, Xiangya School of medicine, The Second Xiangya Hospital, Central South University, Changsha, Hunan, China
| | - Eric Frontera
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Lin Jia
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
| | - Naheed W. Khan
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
| | | | - Thomas A. Ferguson
- Department of Ophthalmology and Visual Sciences, Washington University, St. Louis, MO, USA
| | - Debra A. Thompson
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
- Department of Biological Chemistry, University of Michigan, Ann Arbor, MI, USA
| | - David N. Zacks
- Department of Ophthalmology and Visual Sciences, University of Michigan, Ann Arbor, MI, USA
| |
Collapse
|
117
|
Tsai JC, Miller-Vedam LE, Anand AA, Jaishankar P, Nguyen HC, Renslo AR, Frost A, Walter P. Structure of the nucleotide exchange factor eIF2B reveals mechanism of memory-enhancing molecule. Science 2018; 359:359/6383/eaaq0939. [PMID: 29599213 DOI: 10.1126/science.aaq0939] [Citation(s) in RCA: 123] [Impact Index Per Article: 17.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2017] [Accepted: 02/09/2018] [Indexed: 12/13/2022]
Abstract
Regulation by the integrated stress response (ISR) converges on the phosphorylation of translation initiation factor eIF2 in response to a variety of stresses. Phosphorylation converts eIF2 from a substrate to a competitive inhibitor of its dedicated guanine nucleotide exchange factor, eIF2B, thereby inhibiting translation. ISRIB, a drug-like eIF2B activator, reverses the effects of eIF2 phosphorylation, and in rodents it enhances cognition and corrects cognitive deficits after brain injury. To determine its mechanism of action, we solved an atomic-resolution structure of ISRIB bound in a deep cleft within decameric human eIF2B by cryo-electron microscopy. Formation of fully active, decameric eIF2B holoenzyme depended on the assembly of two identical tetrameric subcomplexes, and ISRIB promoted this step by cross-bridging a central symmetry interface. Thus, regulation of eIF2B assembly emerges as a rheostat for eIF2B activity that tunes translation during the ISR and that can be further modulated by ISRIB.
Collapse
Affiliation(s)
- Jordan C Tsai
- Howard Hughes Medical Institute, University of California, San Francisco, CA, USA.,Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Lakshmi E Miller-Vedam
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA.,Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Aditya A Anand
- Howard Hughes Medical Institute, University of California, San Francisco, CA, USA.,Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| | - Priyadarshini Jaishankar
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Center, University of California, San Francisco, CA, USA
| | - Henry C Nguyen
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA.,Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Adam R Renslo
- Department of Pharmaceutical Chemistry and Small Molecule Discovery Center, University of California, San Francisco, CA, USA
| | - Adam Frost
- Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA. .,Chan Zuckerberg Biohub, San Francisco, CA, USA
| | - Peter Walter
- Howard Hughes Medical Institute, University of California, San Francisco, CA, USA. .,Department of Biochemistry and Biophysics, University of California, San Francisco, CA, USA
| |
Collapse
|
118
|
Wang B, Su CJ, Liu TT, Zhou Y, Feng Y, Huang Y, Liu X, Wang ZH, Chen LH, Luo WF, Liu T. The Neuroprotection of Low-Dose Morphine in Cellular and Animal Models of Parkinson's Disease Through Ameliorating Endoplasmic Reticulum (ER) Stress and Activating Autophagy. Front Mol Neurosci 2018; 11:120. [PMID: 29731707 PMCID: PMC5920031 DOI: 10.3389/fnmol.2018.00120] [Citation(s) in RCA: 39] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2017] [Accepted: 03/28/2018] [Indexed: 12/02/2022] Open
Abstract
Parkinson's disease (PD) is a common neurodegenerative disease characterized the progressive loss of dopaminergic (DA) neurons in the substantia nigra pars compacta (SNc). Brain endogenous morphine biosynthesis was reported to be impaired in PD patients and exogenous morphine attenuated 6-hydroxydopamine (6-OHDA)-induced cell death in vitro. However, the mechanisms underlying neuroprotection of morphine in PD are still unclear. In the present study, we investigated the neuroprotective effects of low-dose morphine in cellular and animal models of PD and the possible underlying mechanisms. Herein, we found 6-OHDA and rotenone decreased the mRNA expression of key enzymes involved in endogenous morphine biosynthesis in SH-SY5Y cells. Incubation of morphine prevented 6-OHDA-induced apoptosis, restored mitochondrial membrane potential, and inhibited the accumulation of intracellular reactive oxygen species (ROS) in SH-SY5Y cells. Furthermore, morphine attenuated the 6-OHDA-induced endoplasmic reticulum (ER) stress possible by activating autophagy in SH-SY5Y cells. Finally, oral application of low-dose morphine significantly improved midbrain tyrosine hydroxylase (TH) expression, decreased apomorphine-evoked rotation and attenuated pain hypersensitivity in a 6-OHDA-induced PD rat model, without the risks associated with morphine addiction. Feeding of low-dose morphine prolonged the lifespan and improved the motor function in several transgenic Drosophila PD models in gender, genotype, and dose-dependent manners. Overall, our results suggest that neuroprotection of low-dose morphine may be mediated by attenuating ER stress and oxidative stress, activating autophagy, and ameliorating mitochondrial function.
Collapse
Affiliation(s)
- Bing Wang
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and the Second Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Cun-Jin Su
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and the Second Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Teng-Teng Liu
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yan Zhou
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Yu Feng
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and the Second Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Ya Huang
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Xu Liu
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Zhi-Hong Wang
- Institute of Neuroscience, Soochow University, Suzhou, China
| | - Li-Hua Chen
- Jiangsu Key Laboratory of Preventive and Translational Medicine for Geriatric Diseases, Department of Nutrition and Food Hygiene, School of Public Health, Soochow University, Suzhou, China
| | - Wei-Feng Luo
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and the Second Affiliated Hospital of Soochow University, Suzhou, China
| | - Tong Liu
- Jiangsu Key Laboratory of Neuropsychiatric Diseases and the Second Affiliated Hospital of Soochow University, Suzhou, China
- Institute of Neuroscience, Soochow University, Suzhou, China
| |
Collapse
|
119
|
Targeting the Endoplasmic Reticulum Unfolded Protein Response to Counteract the Oxidative Stress-Induced Endothelial Dysfunction. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2018; 2018:4946289. [PMID: 29725497 PMCID: PMC5872601 DOI: 10.1155/2018/4946289] [Citation(s) in RCA: 37] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 02/18/2018] [Indexed: 12/22/2022]
Abstract
In endothelial cells, the tight control of the redox environment is essential for the maintenance of vascular homeostasis. The imbalance between ROS production and antioxidant response can induce endothelial dysfunction, the initial event of many cardiovascular diseases. Recent studies have revealed that the endoplasmic reticulum could be a new player in the promotion of the pro- or antioxidative pathways and that in such a modulation, the unfolded protein response (UPR) pathways play an essential role. The UPR consists of a set of conserved signalling pathways evolved to restore the proteostasis during protein misfolding within the endoplasmic reticulum. Although the first outcome of the UPR pathways is the promotion of an adaptive response, the persistent activation of UPR leads to increased oxidative stress and cell death. This molecular switch has been correlated to the onset or to the exacerbation of the endothelial dysfunction in cardiovascular diseases. In this review, we highlight the multiple chances of the UPR to induce or ameliorate oxidative disturbances and propose the UPR pathways as a new therapeutic target for the clinical management of endothelial dysfunction.
Collapse
|
120
|
Imbriani P, Schirinzi T, Meringolo M, Mercuri NB, Pisani A. Centrality of Early Synaptopathy in Parkinson's Disease. Front Neurol 2018; 9:103. [PMID: 29545770 PMCID: PMC5837972 DOI: 10.3389/fneur.2018.00103] [Citation(s) in RCA: 40] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 02/13/2018] [Indexed: 12/16/2022] Open
Abstract
Significant advances have been made in the understanding of the numerous mechanisms involved in Parkinson’s disease (PD) pathogenesis. The identification of PD pathogenic mutations and the use of different animal models have contributed to better elucidate the processes underlying the disease. Here, we report a brief survey of some relevant cellular mechanisms, including autophagic–lysosomal dysfunction, endoplasmic reticulum stress, and mitochondrial impairment, with the main aim to focus on their potential convergent roles in determining early alterations at the synaptic level, mainly consisting in a decrease in dopamine release at nigrostriatal terminals and loss of synaptic plasticity at corticostriatal synapses. In a number of experimental models, this synaptopathy has been shown to be an initial, central event in PD pathogenesis, preceding neuronal damage, thereby representing a valuable tool for testing potential disease-modifying treatments.
Collapse
Affiliation(s)
- Paola Imbriani
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.,Laboratory of Neurophysiology and Plasticity, Fondazione Santa Lucia (IRCCS), Rome, Italy
| | - Tommaso Schirinzi
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.,Laboratory of Neurophysiology and Plasticity, Fondazione Santa Lucia (IRCCS), Rome, Italy
| | - Maria Meringolo
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.,Laboratory of Neurophysiology and Plasticity, Fondazione Santa Lucia (IRCCS), Rome, Italy
| | - Nicola B Mercuri
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.,Laboratory of Neurophysiology and Plasticity, Fondazione Santa Lucia (IRCCS), Rome, Italy
| | - Antonio Pisani
- Department of Systems Medicine, University of Rome "Tor Vergata", Rome, Italy.,Laboratory of Neurophysiology and Plasticity, Fondazione Santa Lucia (IRCCS), Rome, Italy
| |
Collapse
|
121
|
Abstract
The endoplasmic reticulum (ER) is a morphologically dynamic organelle containing different membrane subdomains with distinct cellular functions. Numerous observations have revealed that ER stress response induced by disturbed ER homeostasis is linked to various neurological/neurodegenerative disorders. In contrast, recent findings unveil that ER structural derangements are linked to the progression of several neurological diseases. The derangements involve two distinct, and likely opposing pathways. One is dysfunction of ER dynamics machinery, leading to disruption of ER network organization. Another one is facilitation of pre-existing machinery, leading to generation of markedly-ordered de novo membranous structure. Restoring the ER network can be the effective way toward the cure of ER-deranged neurological disorders.
Collapse
Affiliation(s)
- Tomoyuki Yamanaka
- Laboratory of Structural Neuropathology, Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| | - Nobuyuki Nukina
- Laboratory of Structural Neuropathology, Graduate School of Brain Science, Doshisha University, Kyoto, Japan
| |
Collapse
|
122
|
Rescue of Learning and Memory Deficits in the Human Nonsyndromic Intellectual Disability Cereblon Knock-Out Mouse Model by Targeting the AMP-Activated Protein Kinase-mTORC1 Translational Pathway. J Neurosci 2018; 38:2780-2795. [PMID: 29459374 DOI: 10.1523/jneurosci.0599-17.2018] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/25/2017] [Revised: 01/03/2018] [Accepted: 01/27/2018] [Indexed: 01/05/2023] Open
Abstract
A homozygous nonsense mutation in the cereblon (CRBN) gene results in autosomal recessive, nonsyndromic intellectual disability that is devoid of other phenotypic features, suggesting a critical role of CRBN in mediating learning and memory. In this study, we demonstrate that adult male Crbn knock-out (CrbnKO) mice exhibit deficits in hippocampal-dependent learning and memory tasks that are recapitulated by focal knock-out of Crbn in the adult dorsal hippocampus, with no changes in social or repetitive behavior. Cellular studies identify deficits in long-term potentiation at Schaffer collateral CA1 synapses. We further show that Crbn is robustly expressed in the mouse hippocampus and CrbnKO mice exhibit hyperphosphorylated levels of AMPKα (Thr172). Examination of processes downstream of AMP-activated protein kinase (AMPK) finds that CrbnKO mice have a selective impairment in mediators of the mTORC1 translation initiation pathway in parallel with lower protein levels of postsynaptic density glutamatergic proteins and higher levels of excitatory presynaptic markers in the hippocampus with no change in markers of the unfolded protein response or autophagy pathways. Acute pharmacological inhibition of AMPK activity in adult CrbnKO mice rescues learning and memory deficits and normalizes hippocampal mTORC1 activity and postsynaptic glutamatergic proteins without altering excitatory presynaptic markers. Thus, this study identifies that loss of Crbn results in learning, memory, and synaptic defects as a consequence of exaggerated AMPK activity, inhibition of mTORC1 signaling, and decreased glutamatergic synaptic proteins. Thus, CrbnKO mice serve as an ideal model of intellectual disability to further explore molecular mechanisms of learning and memory.SIGNIFICANCE STATEMENT Intellectual disability (ID) is one of the most common neurodevelopmental disorders. The cereblon (CRBN) gene has been linked to autosomal recessive, nonsyndromic ID, characterized by an intelligence quotient between 50 and 70 but devoid of other phenotypic features, making cereblon an ideal protein for the study of the fundamental aspects of learning and memory. Here, using the cereblon knock-out mouse model, we show that cereblon deficiency disrupts learning, memory, and synaptic function via AMP-activated protein kinase hyperactivity, downregulation of mTORC1, and dysregulation of excitatory synapses, with no changes in social or repetitive behaviors, consistent with findings in the human population. This establishes the cereblon knock-out mouse as a model of pure ID without the confounding behavioral phenotypes associated with other current models of ID.
Collapse
|
123
|
Ting HC, Chang CY, Lu KY, Chuang HM, Tsai SF, Huang MH, Liu CA, Lin SZ, Harn HJ. Targeting Cellular Stress Mechanisms and Metabolic Homeostasis by Chinese Herbal Drugs for Neuroprotection. Molecules 2018; 23:E259. [PMID: 29382106 PMCID: PMC6017457 DOI: 10.3390/molecules23020259] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2017] [Revised: 01/25/2018] [Accepted: 01/26/2018] [Indexed: 12/14/2022] Open
Abstract
Traditional Chinese medicine has been practiced for centuries in East Asia. Herbs are used to maintain health and cure disease. Certain Chinese herbs are known to protect and improve the brain, memory, and nervous system. To apply ancient knowledge to modern science, some major natural therapeutic compounds in herbs were extracted and evaluated in recent decades. Emerging studies have shown that herbal compounds have neuroprotective effects or can ameliorate neurodegenerative diseases. To understand the mechanisms of herbal compounds that protect against neurodegenerative diseases, we summarize studies that discovered neuroprotection by herbal compounds and compound-related mechanisms in neurodegenerative disease models. Those compounds discussed herein show neuroprotection through different mechanisms, such as cytokine regulation, autophagy, endoplasmic reticulum (ER) stress, glucose metabolism, and synaptic function. The interleukin (IL)-1β and tumor necrosis factor (TNF)-α signaling pathways are inhibited by some compounds, thus attenuating the inflammatory response and protecting neurons from cell death. As to autophagy regulation, herbal compounds show opposite regulatory effects in different neurodegenerative models. Herbal compounds that inhibit ER stress prevent neuronal death in neurodegenerative diseases. Moreover, there are compounds that protect against neuronal death by affecting glucose metabolism and synaptic function. Since the progression of neurodegenerative diseases is complicated, and compound-related mechanisms for neuroprotection differ, therapeutic strategies may need to involve multiple compounds and consider the type and stage of neurodegenerative diseases.
Collapse
Affiliation(s)
- Hsiao-Chien Ting
- Bio-innovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (H.-C.T.); (C.-Y.C.); (K.-Y.L.); (H.-M.C.); (M.-H.H.); (C.-A.L.)
| | - Chia-Yu Chang
- Bio-innovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (H.-C.T.); (C.-Y.C.); (K.-Y.L.); (H.-M.C.); (M.-H.H.); (C.-A.L.)
- Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan
| | - Kang-Yun Lu
- Bio-innovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (H.-C.T.); (C.-Y.C.); (K.-Y.L.); (H.-M.C.); (M.-H.H.); (C.-A.L.)
- Graduate Institute of Basic Medical Science, China Medical University, Taichung 404, Taiwan
| | - Hong-Meng Chuang
- Bio-innovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (H.-C.T.); (C.-Y.C.); (K.-Y.L.); (H.-M.C.); (M.-H.H.); (C.-A.L.)
- Agricultural Biotechnology Center, Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan
| | - Sheng-Feng Tsai
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan;
| | - Mao-Hsuan Huang
- Bio-innovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (H.-C.T.); (C.-Y.C.); (K.-Y.L.); (H.-M.C.); (M.-H.H.); (C.-A.L.)
- Department of Life Sciences, National Chung Hsing University, Taichung 402, Taiwan;
| | - Ching-Ann Liu
- Bio-innovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (H.-C.T.); (C.-Y.C.); (K.-Y.L.); (H.-M.C.); (M.-H.H.); (C.-A.L.)
- Department of Medical Research, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan
| | - Shinn-Zong Lin
- Bio-innovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (H.-C.T.); (C.-Y.C.); (K.-Y.L.); (H.-M.C.); (M.-H.H.); (C.-A.L.)
- Department of Neurosurgery, Buddhist Tzu Chi General Hospital, Hualien 970, Taiwan
| | - Horng-Jyh Harn
- Bio-innovation Center, Buddhist Tzu Chi Medical Foundation, Hualien 970, Taiwan; (H.-C.T.); (C.-Y.C.); (K.-Y.L.); (H.-M.C.); (M.-H.H.); (C.-A.L.)
- Department of Pathology, Buddhist Tzu Chi General Hospital and Tzu Chi University, Hualien 970, Taiwan
| |
Collapse
|
124
|
Shih YT, Hsueh YP. The involvement of endoplasmic reticulum formation and protein synthesis efficiency in VCP- and ATL1-related neurological disorders. J Biomed Sci 2018; 25:2. [PMID: 29310658 PMCID: PMC5757295 DOI: 10.1186/s12929-017-0403-3] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2017] [Accepted: 12/26/2017] [Indexed: 12/13/2022] Open
Abstract
The endoplasmic reticulum (ER) is the biggest organelle in cells and is involved in versatile cellular processes. Formation and maintenance of ER morphology are regulated by a series of proteins controlling membrane fusion and curvature. At least six different ER morphology regulators have been demonstrated to be involved in neurological disorders-including Valosin-containing protein (VCP), Atlastin-1 (ATL1), Spastin (SPAST), Reticulon 2 (RTN2), Receptor expression enhancing protein 1 (REEP1) and RAB10-suggesting a critical role of ER formation in neuronal activity and function. Among these genes, mutations in VCP gene involve in inclusion body myopathy with Paget disease of bone and frontotemporal dementia (IBMPFD), familial amyotrophic lateral sclerosis (ALS), autism spectrum disorders (ASD), and hereditary spastic paraplegia (HSP). ATL1 is also one of causative genes of HSP. RAB10 is associated with Parkinson's disease (PD). A recent study showed that VCP and ATL1 work together to regulate dendritic spine formation by controlling ER formation and consequent protein synthesis efficiency. RAB10 shares the same function with VCP and ATL1 to control ER formation and protein synthesis efficiency but acts independently. Increased protein synthesis by adding extra leucine to cultured neurons ameliorated dendritic spine deficits caused by VCP and ATL1 deficiencies, strengthening the significance of protein synthesis in VCP- and ATL1-regulated dendritic spine formation. These findings provide new insight into the roles of ER and protein synthesis in controlling dendritic spine formation and suggest a potential etiology of neurodegenerative disorders caused by mutations in VCP, ATL1 and other genes encoding proteins regulating ER formation and morphogenesis.
Collapse
Affiliation(s)
- Yu-Tzu Shih
- Institute of Molecular Biology, Academia Sinica, 128, Academia Rd., Sec. 2, Taipei, 11529, Taiwan
| | - Yi-Ping Hsueh
- Institute of Molecular Biology, Academia Sinica, 128, Academia Rd., Sec. 2, Taipei, 11529, Taiwan.
| |
Collapse
|
125
|
Athanasiou D, Aguila M, Bellingham J, Li W, McCulley C, Reeves PJ, Cheetham ME. The molecular and cellular basis of rhodopsin retinitis pigmentosa reveals potential strategies for therapy. Prog Retin Eye Res 2018; 62:1-23. [PMID: 29042326 PMCID: PMC5779616 DOI: 10.1016/j.preteyeres.2017.10.002] [Citation(s) in RCA: 239] [Impact Index Per Article: 34.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/03/2017] [Accepted: 10/13/2017] [Indexed: 12/12/2022]
Abstract
Inherited mutations in the rod visual pigment, rhodopsin, cause the degenerative blinding condition, retinitis pigmentosa (RP). Over 150 different mutations in rhodopsin have been identified and, collectively, they are the most common cause of autosomal dominant RP (adRP). Mutations in rhodopsin are also associated with dominant congenital stationary night blindness (adCSNB) and, less frequently, recessive RP (arRP). Recessive RP is usually associated with loss of rhodopsin function, whereas the dominant conditions are a consequence of gain of function and/or dominant negative activity. The in-depth characterisation of many rhodopsin mutations has revealed that there are distinct consequences on the protein structure and function associated with different mutations. Here we categorise rhodopsin mutations into seven discrete classes; with defects ranging from misfolding and disruption of proteostasis, through mislocalisation and disrupted intracellular traffic to instability and altered function. Rhodopsin adRP offers a unique paradigm to understand how disturbances in photoreceptor homeostasis can lead to neuronal cell death. Furthermore, a wide range of therapies have been tested in rhodopsin RP, from gene therapy and gene editing to pharmacological interventions. The understanding of the disease mechanisms associated with rhodopsin RP and the development of targeted therapies offer the potential of treatment for this currently untreatable neurodegeneration.
Collapse
Affiliation(s)
| | - Monica Aguila
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - James Bellingham
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Wenwen Li
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Caroline McCulley
- UCL Institute of Ophthalmology, 11-43 Bath Street, London EC1V 9EL, UK
| | - Philip J Reeves
- School of Biological Sciences, University of Essex, Wivenhoe Park, Essex CO4 3SQ, UK.
| | | |
Collapse
|
126
|
Lucke-Wold B, Seidel K, Udo R, Omalu B, Ornstein M, Nolan R, Rosen C, Ross J. Role of Tau Acetylation in Alzheimer's Disease and Chronic Traumatic Encephalopathy: The Way Forward for Successful Treatment. JOURNAL OF NEUROLOGY AND NEUROSURGERY 2017; 4. [PMID: 29276758 PMCID: PMC5738035] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Progressive neurodegenerative diseases plague millions of individuals both in the United States and across the world. The current pathology of progressive neurodegenerative tauopathies, such as Alzheimer's disease (AD), Pick's disease, frontotemporal dementia (FTD), and progressive supranuclear palsy, primarily revolves around phosphorylation and hyperphosphorylation of the tau protein. However, more recent evidence suggests acetylation of tau protein at lysine 280 may be a critical step in molecular pathology of these neurodegenerative diseases prior to the tau hyperphosphorylation. Secondary injury cascades such as oxidative stress, endoplasmic reticulum stress, and neuroinflammation contribute to lasting damage within the brain and can be induced by a number of different risk factors. These injury cascades funnel into a common pathway of early tau acetylation, which may serve as the catalyst for progressive degeneration. The post translational modification of tau can result in production of toxic oligomers, contributing to reduced solubility as well as aggregation and formation of neurofibrillary tangles, the hallmark of AD pathology. Chronic Traumatic Encephalopathy (CTE), caused by repetitive brain trauma is also associated with a hyperphosphorylation of tau. We postulated acetylation of tau at lysine 280 in CTE disease could be present prior to the hyperphosphorylation and tested this hypothesis in CTE pathologic specimens. We also tested for ac-tau 280 in early stage Alzheimer's disease (Braak stage 1). Histopathological examination using the ac tau 280 antibody was performed in three Alzheimer's cases and three CTE patients. Presence of ac-tau 280 was confirmed in all cases at early sites of disease manifestation. These findings suggest that tau acetylation may precede tau phosphorylation and could be the first "triggering" event leading to neuronal loss. To the best of our knowledge, this is the first study to identify acetylation of the tau protein in CTE. Prevention of tau acetylation could possibly serve as a novel target for stopping neurodegeneration before it fully begins. In this study, we highlight what is known about tau acetylation and neurodegeneration.
Collapse
Affiliation(s)
- Brandon Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV
| | - Kay Seidel
- Dr. Senckenberg Chronomedical Institute, J. W. Goethe University, Frankfurt am Main, Germany
| | - Rub Udo
- Dr. Senckenberg Chronomedical Institute, J. W. Goethe University, Frankfurt am Main, Germany
| | - Bennet Omalu
- Department of Pathology, University of California Davis Medical Center, Davis, CA
| | | | - Richard Nolan
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV
| | - Charles Rosen
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV
| | - Joel Ross
- Cogwellin LLC 4 Industrial Way W, Eatontown NJ, USA
| |
Collapse
|
127
|
Pitale PM, Gorbatyuk O, Gorbatyuk M. Neurodegeneration: Keeping ATF4 on a Tight Leash. Front Cell Neurosci 2017; 11:410. [PMID: 29326555 PMCID: PMC5736573 DOI: 10.3389/fncel.2017.00410] [Citation(s) in RCA: 55] [Impact Index Per Article: 6.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2017] [Accepted: 12/05/2017] [Indexed: 12/15/2022] Open
Abstract
Activation of the endoplasmic reticulum (ER) stress and ER stress response, also known as the unfolded protein response (UPR), is common to various degenerative disorders. Therefore, signaling components of the UPR are currently emerging as potential targets for intervention and treatment of human diseases. One UPR signaling member, activating transcription factor 4 (ATF4), has been found up-regulated in many pathological conditions, pointing to therapeutic potential in targeting its expression. In cells, ATF4 governs multiple signaling pathways, including autophagy, oxidative stress, inflammation, and translation, suggesting a multifaceted role of ATF4 in the progression of various pathologies. However, ATF4 has been shown to trigger both pro-survival and pro-death pathways, and this, perhaps, can explain the contradictory opinions in current literature regarding targeting ATF4 for clinical application. In this review, we summarized recent published studies from our labs and others that focus on the therapeutic potential of the strategy controlling ATF4 expression in different retinal and neurodegenerative disorders.
Collapse
Affiliation(s)
- Priyamvada M Pitale
- Department of Optometry and Vision Science, School of Optometry, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Oleg Gorbatyuk
- Department of Optometry and Vision Science, School of Optometry, University of Alabama at Birmingham, Birmingham, AL, United States.,Department of Neurology, Center for Neurodegeneration and Experimental Therapeutics, University of Alabama at Birmingham, Birmingham, AL, United States
| | - Marina Gorbatyuk
- Department of Optometry and Vision Science, School of Optometry, University of Alabama at Birmingham, Birmingham, AL, United States
| |
Collapse
|
128
|
Litim N, Morissette M, Caruso D, Melcangi RC, Di Paolo T. Effect of the 5α-reductase enzyme inhibitor dutasteride in the brain of intact and parkinsonian mice. J Steroid Biochem Mol Biol 2017; 174:242-256. [PMID: 28982631 DOI: 10.1016/j.jsbmb.2017.09.021] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 09/08/2017] [Accepted: 09/30/2017] [Indexed: 12/15/2022]
Abstract
Dutasteride is a 5alpha-reductase inhibitor in clinical use to treat endocrine conditions. The present study investigated the neuroprotective mechanisms of action of dutasteride in intact and 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP)-lesioned mice using a low dose of MPTP not affecting motor activity modeling early stages of Parkinson's disease (PD). We hypothesized that dutasteride neuroprotection is due to altered steroids levels. Dutasteride pre-treatment prevented loss of striatal dopamine (DA) and its metabolite DOPAC. Dutasteride decreased effects of MPTP on striatal dopamine transporter (DAT), vesicular monoamine transporter 2 (VMAT2) and D2 DA receptor specific binding while D1 receptor specific binding remained unchanged. Dutasteride enhanced DAT specific binding and the glycosylated form of DAT in intact mice. MPTP-lesioned mice had plasma and brain testosterone and dihydrotestosterone levels lower than control mice whereas progesterone and its metabolites (dihydroprogesterone, isopregnanolone and tetrahydroprogesterone) pathway showed increases. Dutasteride treatment by inhibiting transformation of progesterone and testosterone to its metabolites elevated plasma and brain concentrations of testosterone compared to MPTP mice and decreased DHT levels in intact mice. Plasma and brain estradiol levels were low and remained unchanged by MPTP and/or dutasteride treatment. Dutasteride treatment did not affect striatal phosphorylation of Akt and its downstream substrate GSK3β as well as phosphorylation of ERK1/2 in intact and MPTP lesioned MPTP mice. Striatal glial fibrillary acidic protein (GFAP) levels were markedly elevated in MPTP compared to control mice and dutasteride reduced GFAP levels in MPTP mice. Treatment with dutasteride post-lesion left unchanged striatal DA levels. These results suggest dutasteride as promising drug for PD neuroprotection.
Collapse
Affiliation(s)
- Nadhir Litim
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, Canada; Faculty of Pharmacy, Laval University, Quebec City, Canada
| | - Marc Morissette
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, Canada
| | - Donatella Caruso
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Roberto C Melcangi
- Department of Pharmacological and Biomolecular Sciences, Center of Excellence on Neurodegenerative Diseases, Università degli Studi di Milano, Milan, Italy
| | - Thérèse Di Paolo
- Neuroscience Research Unit, Centre Hospitalier Universitaire de Québec, CHUL, Quebec City, Canada; Faculty of Pharmacy, Laval University, Quebec City, Canada.
| |
Collapse
|
129
|
Role of BAG3 in cancer progression: A therapeutic opportunity. Semin Cell Dev Biol 2017; 78:85-92. [PMID: 28864347 DOI: 10.1016/j.semcdb.2017.08.049] [Citation(s) in RCA: 61] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2017] [Revised: 08/26/2017] [Accepted: 08/28/2017] [Indexed: 02/06/2023]
Abstract
BAG3 is a multifunctional protein that can bind to heat shock proteins (Hsp) 70 through its BAG domain and to other partners through its WW domain, proline-rich (PXXP) repeat and IPV (Ile-Pro-Val) motifs. Its intracellular expression can be induced by stressful stimuli, while is constitutive in skeletal muscle, cardiac myocytes and several tumour types. BAG3 can modulate the levels, localisation or activity of its partner proteins, thereby regulating major cell pathways and functions, including apoptosis, autophagy, mechanotransduction, cytoskeleton organisation, motility. A secreted form of BAG3 has been identified in studies on pancreatic ductal adenocarcinoma (PDAC). Secreted BAG3 can bind to a specific receptor, IFITM2, expressed on macrophages, and induce the release of factors that sustain tumour growth and the metastatic process. BAG3 neutralisation therefore appears to constitute a novel potential strategy in the therapy of PDAC and, possibly, other tumours.
Collapse
|