1
|
Wu Y, Xu Q, Chen Y, Li C, Wu Y, Yu X, Li H, Xu Z, Xu J, Ni Z, Ge Y, Yan T, Qi Z, Liu J. Mechanosensitive and pH-Gated Butterfly-Shaped Artificial Ion Channel for High-Selective K + Transport and Cancer Cell Apoptosis. ADVANCED MATERIALS (DEERFIELD BEACH, FLA.) 2025; 37:e2416852. [PMID: 39981913 DOI: 10.1002/adma.202416852] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2024] [Revised: 02/10/2025] [Indexed: 02/22/2025]
Abstract
To advance the exploration of mechanisms underlying natural multi-gated ion channels, a novel butterfly-shaped biomimetic K+ channel GnC7 (n = 3, 4) is developed with dual mechanical and pH responsiveness, exhibiting unprecedented K+/Na+ selectivity (G3C7: 34.4; G4C7: 41.3). These channels constructed from poly(propylene imine) dendrimer and benzo-21-crown-7-ethers achieve high K+ transport activity (EC50: 0.72 µm for G3C7; 0.9 µm for G4C7) due to their arc-like mechanical rotation. The dynamic mode relies on butterfly-shaped topology derived from the highly symmetrical core and multiple intramolecular hydrogen bonds. GnC7 can sense mechanical stimulus applied to liposomes/cells and then adapt the K+ transport rate accordingly. Furthermore, reversible ON/OFF switching of K+ transport is realized through the pH-controllable host-guest complexation. G4C7-induced ultrafast cellular K+ efflux (70% within only 9 min) efficiently triggers mitochondrial-dependent apoptosis of cancer cells by provoking endoplasmic reticulum stress accompanied by drastic Ca2+ sparks. This work embodies a multi-dimensional regulation of channel functions; it will provide insights into the dynamic behaviors of biological analogs and promote the innovative design of artificial ion channels and therapeutic agents.
Collapse
Affiliation(s)
- Yaqi Wu
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an, 710129, China
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, China
| | - Qiangqiang Xu
- Sino-German Joint Research Lab for Space Biomaterials and Translational Technology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Yaoxuan Chen
- Sino-German Joint Research Lab for Space Biomaterials and Translational Technology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Cong Li
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an, 710129, China
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, China
| | - Yanliang Wu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, China
| | - Xiaoxuan Yu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, China
- Sino-German Joint Research Lab for Space Biomaterials and Translational Technology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Hui Li
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, China
- Sino-German Joint Research Lab for Space Biomaterials and Translational Technology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Zhengwei Xu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, China
| | - Jiayun Xu
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, China
| | - Zhigang Ni
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, China
| | - Yan Ge
- Sino-German Joint Research Lab for Space Biomaterials and Translational Technology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Tengfei Yan
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, China
| | - Zhenhui Qi
- Sino-German Joint Research Lab for Space Biomaterials and Translational Technology, School of Life Sciences, Northwestern Polytechnical University, Xi'an, 710072, China
| | - Junqiu Liu
- College of Chemistry and Chemical Engineering, Key Laboratory of Special Functional and Smart Polymer Materials of Ministry of Industry and Information Technology, Northwestern Polytechnical University, Xi'an, 710129, China
- College of Material, Chemistry and Chemical Engineering, Key Laboratory of Organosilicon Chemistry and Material Technology, Ministry of Education, Hangzhou Normal University, Hangzhou, 311121, China
| |
Collapse
|
2
|
Dossat AM, Trychta KA, Glotfelty EJ, Hinkle JJ, Fortuno LV, Gore LN, Richie CT, Harvey BK. Excitotoxic glutamate levels cause the secretion of resident endoplasmic reticulum proteins. J Neurochem 2024; 168:2461-2478. [PMID: 38491746 PMCID: PMC11401966 DOI: 10.1111/jnc.16093] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2023] [Revised: 02/09/2024] [Accepted: 02/23/2024] [Indexed: 03/18/2024]
Abstract
Dysregulation of synaptic glutamate levels can lead to excitotoxicity such as that observed in stroke, traumatic brain injury, and epilepsy. The role of increased intracellular calcium (Ca2+) in the development of excitotoxicity is well established. However, less is known regarding the impact of glutamate on endoplasmic reticulum (ER)-Ca2+-mediated processes such as proteostasis. To investigate this, we expressed a secreted ER Ca2+ modulated protein (SERCaMP) in primary cortical neurons to monitor exodosis, a phenomenon whereby ER calcium depletion causes the secretion of ER-resident proteins that perform essential functions to the ER and the cell. Activation of glutamatergic receptors (GluRs) led to an increase in SERCaMP secretion indicating that normally ER-resident proteins are being secreted in a manner consistent with ER Ca2+ depletion. Antagonism of ER Ca2+ channels attenuated the effects of glutamate and GluR agonists on SERCaMP release. We also demonstrate that endogenous proteins containing an ER retention/retrieval sequence (ERS) are secreted in response to GluR activation supporting that neuronal activation by glutamate promotes ER exodosis. Ectopic expression of KDEL receptors attenuated the secretion of ERS-containing proteins caused by GluR agonists. Taken together, our data indicate that excessive GluR activation causes disruption of neuronal proteostasis by triggering the secretion of ER-resident proteins through ER Ca2+ depletion and describes a new facet of excitotoxicity.
Collapse
Affiliation(s)
- Amanda M. Dossat
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, 21224
| | - Kathleen A. Trychta
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, 21224
| | - Elliot J. Glotfelty
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, 21224
| | - Joshua J. Hinkle
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, 21224
| | - Lowella V. Fortuno
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, 21224
| | - Lana N. Gore
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, 21224
| | - Christopher T. Richie
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, 21224
| | - Brandon K. Harvey
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, 21224
| |
Collapse
|
3
|
Parkkinen I, Their A, Asghar MY, Sree S, Jokitalo E, Airavaara M. Pharmacological Regulation of Endoplasmic Reticulum Structure and Calcium Dynamics: Importance for Neurodegenerative Diseases. Pharmacol Rev 2023; 75:959-978. [PMID: 37127349 DOI: 10.1124/pharmrev.122.000701] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2022] [Revised: 03/27/2023] [Accepted: 04/04/2023] [Indexed: 05/03/2023] Open
Abstract
The endoplasmic reticulum (ER) is the largest organelle of the cell, composed of a continuous network of sheets and tubules, and is involved in protein, calcium (Ca2+), and lipid homeostasis. In neurons, the ER extends throughout the cell, both somal and axodendritic compartments, and is highly important for neuronal functions. A third of the proteome of a cell, secreted and membrane-bound proteins, are processed within the ER lumen and most of these proteins are vital for neuronal activity. The brain itself is high in lipid content, and many structural lipids are produced, in part, by the ER. Cholesterol and steroid synthesis are strictly regulated in the ER of the blood-brain barrier protected brain cells. The high Ca2+ level in the ER lumen and low cytosolic concentration is needed for Ca2+-based intracellular signaling, for synaptic signaling and Ca2+ waves, and for preparing proteins for correct folding in the presence of high Ca2+ concentrations to cope with the high concentrations of extracellular milieu. Particularly, ER Ca2+ is controlled in axodendritic areas for proper neurito- and synaptogenesis and synaptic plasticity and remodeling. In this review, we cover the physiologic functions of the neuronal ER and discuss it in context of common neurodegenerative diseases, focusing on pharmacological regulation of ER Ca2+ Furthermore, we postulate that heterogeneity of the ER, its protein folding capacity, and ensuring Ca2+ regulation are crucial factors for the aging and selective vulnerability of neurons in various neurodegenerative diseases. SIGNIFICANCE STATEMENT: Endoplasmic reticulum (ER) Ca2+ regulators are promising therapeutic targets for degenerative diseases for which efficacious drug therapies do not exist. The use of pharmacological probes targeting maintenance and restoration of ER Ca2+ can provide restoration of protein homeostasis (e.g., folding of complex plasma membrane signaling receptors) and slow down the degeneration process of neurons.
Collapse
Affiliation(s)
- Ilmari Parkkinen
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Anna Their
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Muhammad Yasir Asghar
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Sreesha Sree
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Eija Jokitalo
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- Neuroscience Center (I.P., A.T., M.A.), Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy (I.P., M.A.), Cell and Tissue Dynamics Research Program, Institute of Biotechnology, Helsinki Institute of Life Sciences (M.Y.A., S.S., E.J.), and Electron Microscopy Unit, Institute of Biotechnology, Helsinki Institute of Life Sciences (E.J.), University of Helsinki, Helsinki, Finland
| |
Collapse
|
4
|
Svarcbahs R, Blossom SM, Baffoe-Bonnie HS, Trychta KA, Greer LK, Pickel J, Henderson MJ, Harvey BK. A transgenic mouse line for assaying tissue-specific changes in endoplasmic reticulum proteostasis. Transgenic Res 2023; 32:209-221. [PMID: 37133648 PMCID: PMC10195735 DOI: 10.1007/s11248-023-00349-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/19/2022] [Accepted: 04/17/2023] [Indexed: 05/04/2023]
Abstract
Maintenance of calcium homeostasis is important for proper endoplasmic reticulum (ER) function. When cellular stress conditions deplete the high concentration of calcium in the ER, ER-resident proteins are secreted into the extracellular space in a process called exodosis. Monitoring exodosis provides insight into changes in ER homeostasis and proteostasis resulting from cellular stress associated with ER calcium dysregulation. To monitor cell-type specific exodosis in the intact animal, we created a transgenic mouse line with a Gaussia luciferase (GLuc)-based, secreted ER calcium-modulated protein, SERCaMP, preceded by a LoxP-STOP-LoxP (LSL) sequence. The Cre-dependent LSL-SERCaMP mice were crossed with albumin (Alb)-Cre and dopamine transporter (DAT)-Cre mouse lines. GLuc-SERCaMP expression was characterized in mouse organs and extracellular fluids, and the secretion of GLuc-SERCaMP in response to cellular stress was monitored following pharmacological depletion of ER calcium. In LSL-SERCaMP × Alb-Cre mice, robust GLuc activity was observed only in the liver and blood, whereas in LSL-SERCaMP × DAT-Cre mice, GLuc activity was seen in midbrain dopaminergic neurons and tissue samples innervated by dopaminergic projections. After calcium depletion, we saw increased GLuc signal in the plasma and cerebrospinal fluid collected from the Alb-Cre and DAT-Cre crosses, respectively. This mouse model can be used to investigate the secretion of ER-resident proteins from specific cell and tissue types during disease pathogenesis and may aid in the identification of therapeutics and biomarkers of disease.
Collapse
Affiliation(s)
- Reinis Svarcbahs
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Sarah M Blossom
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Helena S Baffoe-Bonnie
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Kathleen A Trychta
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Lacey K Greer
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
| | - James Pickel
- Transgenic Technology Core, Intramural Research Program, National Institute of Mental Health, Bethesda, MD, 20892, USA
| | - Mark J Henderson
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD, 20850, USA
| | - Brandon K Harvey
- Cellular Stress and Inflammation Section, Intramural Research Program, National Institute On Drug Abuse, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
5
|
Wu KJ, Hung TW, Wang YS, Chen YH, Bae EK, Yu SJ. Prosaposin PS18 reduces dopaminergic neurodegeneration in a 6-hydroxydopamine rat model of Parkinson's disease. Sci Rep 2023; 13:8148. [PMID: 37208379 DOI: 10.1038/s41598-023-35274-6] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2023] [Accepted: 05/16/2023] [Indexed: 05/21/2023] Open
Abstract
Saposin and its precursor prosaposin are endogenous proteins with neurotrophic and anti-apoptotic properties. Prosaposin or its analog prosaposin-derived 18-mer peptide (PS18) reduced neuronal damage in hippocampus and apoptosis in stroke brain. Its role in Parkinson's disease (PD) has not been well characterized. This study aimed to examine the physiological role of PS18 in 6-hydroxydopamine (6-OHDA) cellular and animal models of PD. We found that PS18 significantly antagonized 6-OHDA -mediated dopaminergic neuronal loss and TUNEL in rat primary dopaminergic neuronal culture. In SH-SY5Y cells overexpressing the secreted ER calcium-monitoring proteins, we found that PS18 significantly reduced thapsigargin and 6-OHDA-mediated ER stress. The expression of prosaposin and the protective effect of PS18 were next examined in hemiparkinsonian rats. 6-OHDA was unilaterally administered to striatum. The expression of prosaposin was transiently upregulated in striatum on D3 (day 3) after lesioning and returned below the basal level on D29. The 6-OHDA-lesioned rats developed bradykinesia and an increase in methamphetamine-mediated rotation, which was antagonized by PS18. Brain tissues were collected for Western blot, immunohistochemistry, and qRTPCR analysis. Tyrosine hydroxylase immunoreactivity was significantly reduced while the expressions of PERK, ATF6, CHOP, and BiP were upregulated in the lesioned nigra; these responses were significantly antagonized by PS18. Taken together, our data support that PS18 is neuroprotective in cellular and animal models of PD. The mechanisms of protection may involve anti-ER stress.
Collapse
Affiliation(s)
- Kuo-Jen Wu
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Miaoli, Taiwan
| | - Tsai-Wei Hung
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Miaoli, Taiwan
| | - Yu-Syuan Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Miaoli, Taiwan
| | - Yun-Hsiang Chen
- Department of Life Science, Fu-Jen Catholic University, New Taipei City, Taiwan
| | - Eun-Kyung Bae
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Miaoli, Taiwan
| | - Seong-Jin Yu
- Center for Neuropsychiatric Research, National Health Research Institutes, 35 Keyan Road, Zhunan, 35053, Miaoli, Taiwan.
| |
Collapse
|
6
|
Protective mechanisms by glial cell line-derived neurotrophic factor and cerebral dopamine neurotrophic factor against the α-synuclein accumulation in Parkinson's disease. Biochem Soc Trans 2023; 51:245-257. [PMID: 36794783 DOI: 10.1042/bst20220770] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/13/2022] [Revised: 01/25/2023] [Accepted: 01/26/2023] [Indexed: 02/17/2023]
Abstract
Synucleinopathies constitute a disease family named after alpha-synuclein protein, which is a significant component of the intracellular inclusions called Lewy bodies. Accompanying the progressive neurodegeneration, Lewy bodies and neurites are the main histopathologies of synucleinopathies. The complicated role of alpha-synuclein in the disease pathology makes it an attractive therapeutic target for disease-modifying treatments. GDNF is one of the most potent neurotrophic factors for dopamine neurons, whereas CDNF is protective and neurorestorative with entirely different mechanisms of action. Both have been in the clinical trials for the most common synucleinopathy, Parkinson's disease. With the AAV-GDNF clinical trials ongoing and the CDNF trial being finalized, their effects on abnormal alpha-synuclein accumulation are of great interest. Previous animal studies with an alpha-synuclein overexpression model have shown that GDNF was ineffective against alpha-synuclein accumulation. However, a recent study with cell culture and animal models of alpha-synuclein fibril inoculation has demonstrated the opposite by revealing that the GDNF/RET signaling cascade is required for the protective effect of GDNF on alpha-synuclein aggregation. CDNF, an ER resident protein, was shown to bind alpha-synuclein directly. CDNF reduced the uptake of alpha-synuclein fibrils by the neurons and alleviated the behavioral deficits induced by fibrils injected into the mouse brain. Thus, GDNF and CDNF can modulate different symptoms and pathologies of Parkinson's disease, and perhaps, similarly for other synucleinopathies. Their unique mechanisms for preventing alpha-synuclein-related pathology should be studied more carefully to develop disease-modifying therapies.
Collapse
|
7
|
Zhang JX, Zhou KG, Yin YX, Jin LJ, Tong WF, Guo J, Yu LH, Ye XC, Jiang M. Mesencephalic astrocyte-derived neurotrophic factor (MANF) prevents the neuroinflammation induced dopaminergic neurodegeneration. Exp Gerontol 2023; 171:112037. [PMID: 36436758 DOI: 10.1016/j.exger.2022.112037] [Citation(s) in RCA: 9] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2022] [Revised: 11/14/2022] [Accepted: 11/21/2022] [Indexed: 11/27/2022]
Abstract
BACKGROUND The excessive activation of the microglia leads to the release of inflammatory factors that contribute to neuronal cell loss and neurodegeneration in Parkinson's Disease (PD). Mesencephalic astrocyte-derived neurotrophic factor (MANF) that belongs to a newly found neurotrophic factors (NTFs) family has been reported to promote neuronal survival in the PD models. However, the effects of the MANF on neuroinflammation in PD remain unclear. METHODS AAV8-MANF virus was constructed to determine whether the high expression of MANF can protect the neuroinflammation-induced dopaminergic neurodegeneration in rats with 6-OHDA-induced PD. Rotarod performance test, immunofluorescent staining and western bolt were employed to evaluate the behavioral dysfunction, dopaminergic neurodegeneration, microglia activation, and signal activation. 6-OHDA treated SH-SY5Y cells and LPS treated BV-2 cells were used as the in vitro model for MANF neuroprotective and neuroinflammation mechanisms. Cell vitality and apoptosis were evaluated with MTT, CCK-8 and flow cytometric analysis. The AKT/GSK3β-Nrf2 signaling and the TNF-α/IL6 expression were measured by Western Blot. RESULTS Our findings indicated that the elevated MANF expression by the AAV8-MANF administration ameliorated the motor dysfunction and protected the dopaminergic neurons in the 6-OHDA treated rats. The upregulated CD11b in the rat SN caused by the 6-OHDA administration was significantly attenuated by the pretreatment of the AAV8-MANF. Furthermore, the levels of p-AKT, p-GSK3β, BCL-2, and Nrf-2 were upregulated by the high expression of the MANF. Under the oxidative stress of the 6-OHDA, the MANF significantly reduced the apoptotic effect of the TNF-α on the SH-SY5Y cells. In the LPS treated BV-2 cells, the MANF reduced the production of the TNF-α and IL-6, via enhancing the Nrf-2, p-Akt, p-GSK3β, and p-NF-κβ level. CONCLUSIONS These results suggested that the MANF prevented the dopaminergic neurodegeneration caused by the microglia activation in PD via activation of the AKT/GSK3β-Nrf-2 signaling axis.
Collapse
Affiliation(s)
- Jing-Xing Zhang
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Kai-Ge Zhou
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Yan-Xin Yin
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China; Biomedical Research Center, Tongji University Suzhou Institute, Building 2198 Jinfeng Road, Wuzhong District, Suzhou, Jiangsu 215101, China; School of Life Science and Technology, Tongji University, 1239 Siping Road, Shanghai 200092, China
| | - Ling-Jing Jin
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China; Department of Neurology and Neurological Rehabilitation, Shanghai Yangzhi Rehabilitation Hospital, School of Medicine, Tongji University, Shanghai 200092, China; Shanghai Clinical Research Center for Aging and Medicine, Shanghai 200040, China
| | - Wei-Fang Tong
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China
| | - Jia Guo
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China; Biomedical Research Center, Tongji University Suzhou Institute, Building 2198 Jinfeng Road, Wuzhong District, Suzhou, Jiangsu 215101, China
| | - Li-Hua Yu
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China; Biomedical Research Center, Tongji University Suzhou Institute, Building 2198 Jinfeng Road, Wuzhong District, Suzhou, Jiangsu 215101, China
| | - Xian-Cheng Ye
- Department of Pharmacy, Tongji Hospital, Tongji University School of Medicine, Shanghai 200065, China.
| | - Ming Jiang
- Department of Neurology, Tongji Hospital, School of Medicine, Tongji University, Shanghai 200092, China; Biomedical Research Center, Tongji University Suzhou Institute, Building 2198 Jinfeng Road, Wuzhong District, Suzhou, Jiangsu 215101, China.
| |
Collapse
|
8
|
Deng H, Zhang P, Gao X, Chen W, Li J, Wang F, Gu Y, Hou X. Emerging trophic activities of mesencephalic astrocyte-derived neurotrophic factor in tissue repair and regeneration. Int Immunopharmacol 2023; 114:109598. [PMID: 36538855 DOI: 10.1016/j.intimp.2022.109598] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2022] [Revised: 12/05/2022] [Accepted: 12/12/2022] [Indexed: 12/23/2022]
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a soluble endoplasmic reticulum (ER) luminal protein and its expression and secretion can be induced by ER stress. Despite initially being classified as a neurotrophic factor, MANF has been demonstrated to have restorative and protective effects in many different cell types such as neurons, liver cells, retinal cells, cardiac myocytes, and pancreatic β cells. However, underlying molecular mechanisms are complex and remain incompletely understood. The aims of this review are to highlight the latest advances in the understanding of the trophic activities of MANF in tissue repair and regeneration as well as underlying molecular mechanisms. The structural motifs and immune modulation of MANF are also described. We therefore propose that MANF might be a promising therapeutic target for tissue repair.
Collapse
Affiliation(s)
- Haiyan Deng
- School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Pingping Zhang
- Institute of Clinical Pharmacology, Anhui Medical University, Key Laboratory of Anti-inflammatory and Immune Medicine, Ministry of Education, Anhui Collaborative Innovation Center of Anti-inflammatory and Immune Medicine, Hefei, PR China
| | - Xianxian Gao
- School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Weiyi Chen
- School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Jianing Li
- School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, PR China
| | - Fuyan Wang
- School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, PR China; Qingdao Hospital of Traditional Chinese Medicine (Qingdao Hiser Hospital), Qingdao, 266000, PR China
| | - Yiyue Gu
- Department of Cardiology, Xuzhou No.1 Peoples Hospital, Xuzhou, PR China
| | - Xin Hou
- School of Medicine, Ningbo University, Ningbo, Zhejiang 315211, PR China; The Affiliated Hospital of Medical School, Ningbo University, Ningbo, PR China.
| |
Collapse
|
9
|
Liu YY, Huo D, Zeng LT, Fan GQ, Shen T, Zhang TM, Cai JP, Cui J. Mesencephalic astrocyte-derived neurotrophic factor (MANF): Structure, functions and therapeutic potential. Ageing Res Rev 2022; 82:101763. [PMID: 36272696 DOI: 10.1016/j.arr.2022.101763] [Citation(s) in RCA: 19] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2022] [Revised: 09/18/2022] [Accepted: 10/15/2022] [Indexed: 01/31/2023]
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a novel evolutionarily conserved protein present in both vertebrate and invertebrate species. MANF shows distinct structural and functional properties than the traditional neurotrophic factors (NTF). MANF is composed of an N-terminal saposin-like lipid-binding domain and a C-terminal SAF-A/B, Acinus and PIAS (SAP) domain connected by a short linker. The two well-described activities of MANF include (1) role as a neurotrophic factor that plays direct neuroprotective effects in the nervous system and (2) cell protective effects in the animal models of non-neuronal diseases, including retinal damage, diabetes mellitus, liver injury, myocardial infarction, nephrotic syndrome, etc. The main objective of the current review is to provide up-to-date insights regarding the structure of MANF, mechanisms regulating its expression and secretion, physiological functions in various tissues and organs, protective effects during aging, and potential clinical applications. Together, this review highlights the importance of MANF in reversing age-related dysfunction and geroprotection.
Collapse
Affiliation(s)
- Yuan-Yuan Liu
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, PR China
| | - Da Huo
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, PR China
| | - Lv-Tao Zeng
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, PR China
| | - Guo-Qing Fan
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, PR China
| | - Tao Shen
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, PR China
| | - Tie-Mei Zhang
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, PR China
| | - Jian-Ping Cai
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, PR China.
| | - Ju Cui
- The Key Laboratory of Geriatrics, Beijing Institute of Geriatrics, Institute of Geriatric Medicine, Chinese Academy of Medical Science, Beijing Hospital/National Center of Gerontology of National Health Commission, Beijing, PR China.
| |
Collapse
|
10
|
Jäntti MH, Jackson SN, Kuhn J, Parkkinen I, Sree S, Hinkle JJ, Jokitalo E, Deterding LJ, Harvey BK. Palmitate and thapsigargin have contrasting effects on ER membrane lipid composition and ER proteostasis in neuronal cells. Biochim Biophys Acta Mol Cell Biol Lipids 2022; 1867:159219. [PMID: 35981704 PMCID: PMC9452468 DOI: 10.1016/j.bbalip.2022.159219] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2022] [Revised: 07/21/2022] [Accepted: 08/10/2022] [Indexed: 11/25/2022]
Abstract
The endoplasmic reticulum (ER) is an organelle that performs several key functions such as protein synthesis and folding, lipid metabolism and calcium homeostasis. When these functions are disrupted, such as upon protein misfolding, ER stress occurs. ER stress can trigger adaptive responses to restore proper functioning such as activation of the unfolded protein response (UPR). In certain cells, the free fatty acid palmitate has been shown to induce the UPR. Here, we examined the effects of palmitate on UPR gene expression in a human neuronal cell line and compared it with thapsigargin, a known depletor of ER calcium and trigger of the UPR. We used a Gaussia luciferase-based reporter to assess how palmitate treatment affects ER proteostasis and calcium homeostasis in the cells. We also investigated how ER calcium depletion by thapsigargin affects lipid membrane composition by performing mass spectrometry on subcellular fractions and compared this to palmitate. Surprisingly, palmitate treatment did not activate UPR despite prominent changes to membrane phospholipids. Conversely, thapsigargin induced a strong UPR, but did not significantly change the membrane lipid composition in subcellular fractions. In summary, our data demonstrate that changes in membrane lipid composition and disturbances in ER calcium homeostasis have a minimal influence on each other in neuronal cells. These data provide new insight into the adaptive interplay of lipid homeostasis and proteostasis in the cell.
Collapse
Affiliation(s)
- Maria H Jäntti
- Molecular Mechanisms of Cellular Stress and Inflammation, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, USA.
| | - Shelley N Jackson
- Translational Analytical Core, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, USA
| | - Jeffrey Kuhn
- Mass Spectrometry Research and Support Group, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
| | - Ilmari Parkkinen
- Neuroscience Center, Helsinki Institute for Life Science, University of Helsinki, Haartmaninkatu 8, 00014 Helsinki, Finland
| | - Sreesha Sree
- Cell and Tissue Dynamics Research Programme, Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Finland
| | - Joshua J Hinkle
- Molecular Mechanisms of Cellular Stress and Inflammation, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, USA
| | - Eija Jokitalo
- Cell and Tissue Dynamics Research Programme, Institute of Biotechnology, Helsinki Institute of Life Science, University of Helsinki, Finland
| | - Leesa J Deterding
- Mass Spectrometry Research and Support Group, National Institute of Environmental Health Sciences, Research Triangle Park, Durham, NC 27709, USA
| | - Brandon K Harvey
- Molecular Mechanisms of Cellular Stress and Inflammation, Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, USA.
| |
Collapse
|
11
|
Guo D, Huang X, Xiong T, Wang X, Zhang J, Wang Y, Liang J. Molecular mechanisms of programmed cell death in methamphetamine-induced neuronal damage. Front Pharmacol 2022; 13:980340. [PMID: 36059947 PMCID: PMC9428134 DOI: 10.3389/fphar.2022.980340] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2022] [Accepted: 07/28/2022] [Indexed: 12/02/2022] Open
Abstract
Methamphetamine, commonly referred to as METH, is a highly addictive psychostimulant and one of the most commonly misused drugs on the planet. Using METH continuously can increase your risk for drug addiction, along with other health complications like attention deficit disorder, memory loss, and cognitive decline. Neurotoxicity caused by METH is thought to play a significant role in the onset of these neurological complications. The molecular mechanisms responsible for METH-caused neuronal damage are discussed in this review. According to our analysis, METH is closely associated with programmed cell death (PCD) in the process that causes neuronal impairment, such as apoptosis, autophagy, necroptosis, pyroptosis, and ferroptosis. In reviewing this article, some insights are gained into how METH addiction is accompanied by cell death and may help to identify potential therapeutic targets for the neurological impairment caused by METH abuse.
Collapse
Affiliation(s)
- Dongming Guo
- Institute of Translational Medicine, Medical, Yangzhou University, Yangzhou, China
| | - Xinlei Huang
- Institute of Translational Medicine, Medical, Yangzhou University, Yangzhou, China
| | - Tianqing Xiong
- Institute of Translational Medicine, Medical, Yangzhou University, Yangzhou, China
| | - Xingyi Wang
- Institute of Translational Medicine, Medical, Yangzhou University, Yangzhou, China
| | - Jingwen Zhang
- Institute of Translational Medicine, Medical, Yangzhou University, Yangzhou, China
| | - Yingge Wang
- Department of Neurology, Affiliated Hospital of Yangzhou University, Yangzhou, China
| | - Jingyan Liang
- Institute of Translational Medicine, Medical, Yangzhou University, Yangzhou, China
- *Correspondence: Jingyan Liang,
| |
Collapse
|
12
|
Greer LK, Meilleur KG, Harvey BK, Wires ES. Identification of ER/SR resident proteins as biomarkers for ER/SR calcium depletion in skeletal muscle cells. Orphanet J Rare Dis 2022; 17:225. [PMID: 35698232 PMCID: PMC9195201 DOI: 10.1186/s13023-022-02368-9] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/03/2022] [Accepted: 05/29/2022] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Aberrations to endoplasmic/sarcoplasmic reticulum (ER/SR) calcium concentration can result in the departure of endogenous proteins in a phenomenon termed exodosis. Redistribution of the ER/SR proteome can have deleterious effects to cell function and cell viability, often contributing to disease pathogenesis. Many proteins prone to exodosis reside in the ER/SR via an ER retention/retrieval sequence (ERS) and are involved in protein folding, protein modification, and protein trafficking. While the consequences of their extracellular presence have yet to be fully delineated, the proteins that have undergone exodosis may be useful for biomarker development. Skeletal muscle cells rely upon tightly coordinated ER/SR calcium release for muscle contractions, and perturbations to calcium homeostasis can result in myopathies. Ryanodine receptor type-1 (RYR1) is a calcium release channel located in the SR. Mutations to the RYR1 gene can compromise calcium homeostasis leading to a vast range of clinical phenotypes encompassing hypotonia, myalgia, respiratory insufficiency, ophthalmoplegia, fatigue and malignant hyperthermia (MH). There are currently no FDA approved treatments for RYR1-related myopathies (RYR1-RM). RESULTS Here we examine the exodosis profile of skeletal muscle cells following ER/SR calcium depletion. Proteomic analysis identified 4,465 extracellular proteins following ER/SR calcium depletion with 1,280 proteins significantly different than vehicle. A total of 54 ERS proteins were identified and 33 ERS proteins significantly increased following ER/SR calcium depletion. Specifically, ERS protein, mesencephalic astrocyte-derived neurotrophic factor (MANF), was elevated following calcium depletion, making it a potential biomarker candidate for human samples. Despite no significant elevation of MANF in plasma levels among healthy volunteers and RYR1-RM individuals, MANF plasma levels positively correlated with age in RYR1-RM individuals, presenting a potential biomarker of disease progression. Selenoprotein N (SEPN1) was also detected only in extracellular samples following ER/SR calcium depletion. This protein is integral to calcium handling and SEPN1 variants have a causal role in SEPN1-related myopathies (SEPN1-RM). Extracellular presence of ER/SR membrane proteins may provide new insight into proteomic alterations extending beyond ERS proteins. Pre-treatment of skeletal muscle cells with bromocriptine, an FDA approved drug recently found to have anti-exodosis effects, curbed exodosis of ER/SR resident proteins. CONCLUSION Changes to the extracellular content caused by intracellular calcium dysregulation presents an opportunity for biomarker development and drug discovery.
Collapse
Affiliation(s)
- Lacey K Greer
- National Institute On Drug Abuse, 251 Bayview Blvd, Baltimore, MD, 21224, USA
| | | | - Brandon K Harvey
- National Institute On Drug Abuse, 251 Bayview Blvd, Baltimore, MD, 21224, USA.
| | - Emily S Wires
- National Institute On Drug Abuse, 251 Bayview Blvd, Baltimore, MD, 21224, USA.
| |
Collapse
|
13
|
Gansemer ER, Rutkowski DT. Pathways Linking Nicotinamide Adenine Dinucleotide Phosphate Production to Endoplasmic Reticulum Protein Oxidation and Stress. Front Mol Biosci 2022; 9:858142. [PMID: 35601828 PMCID: PMC9114485 DOI: 10.3389/fmolb.2022.858142] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2022] [Accepted: 04/04/2022] [Indexed: 11/13/2022] Open
Abstract
The endoplasmic reticulum (ER) lumen is highly oxidizing compared to other subcellular compartments, and maintaining the appropriate levels of oxidizing and reducing equivalents is essential to ER function. Both protein oxidation itself and other essential ER processes, such as the degradation of misfolded proteins and the sequestration of cellular calcium, are tuned to the ER redox state. Simultaneously, nutrients are oxidized in the cytosol and mitochondria to power ATP generation, reductive biosynthesis, and defense against reactive oxygen species. These parallel needs for protein oxidation in the ER and nutrient oxidation in the cytosol and mitochondria raise the possibility that the two processes compete for electron acceptors, even though they occur in separate cellular compartments. A key molecule central to both processes is NADPH, which is produced by reduction of NADP+ during nutrient catabolism and which in turn drives the reduction of components such as glutathione and thioredoxin that influence the redox potential in the ER lumen. For this reason, NADPH might serve as a mediator linking metabolic activity to ER homeostasis and stress, and represent a novel form of mitochondria-to-ER communication. In this review, we discuss oxidative protein folding in the ER, NADPH generation by the major pathways that mediate it, and ER-localized systems that can link the two processes to connect ER function to metabolic activity.
Collapse
Affiliation(s)
- Erica R. Gansemer
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| | - D. Thomas Rutkowski
- Department of Anatomy and Cell Biology, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
- Department of Internal Medicine, Carver College of Medicine, University of Iowa, Iowa City, IA, United States
| |
Collapse
|
14
|
Abstract
Constitutive vesicle trafficking is the default pathway used by all cells for movement of intracellular cargoes between subcellular compartments and in and out of the cell. Classically, constitutive trafficking was thought to be continuous and unregulated, in contrast to regulated secretion, wherein vesicles are stored intracellularly until undergoing synchronous membrane fusion following a Ca2+ signal. However, as shown in the literature reviewed here, many continuous trafficking steps can be up- or down-regulated by Ca2+, including several steps associated with human pathologies. Notably, we describe a series of Ca2+ pumps, channels, Ca2+-binding effector proteins, and their trafficking machinery targets that together regulate the flux of cargo in response to genetic alterations as well as baseline and agonist-dependent Ca2+ signals. Here, we review the most recent advances, organized by organellar location, that establish the importance of these components in trafficking steps. Ultimately, we conclude that Ca2+ regulates an expanding series of distinct mechanistic steps. Furthermore, the involvement of Ca2+ in trafficking is complex. For example, in some cases, the same Ca2+ effectors regulate surprisingly distinct trafficking steps, or even the same trafficking step with opposing influences, through binding to different target proteins.
Collapse
Affiliation(s)
- John Sargeant
- Division of Biological Sciences & Center for Structural & Functional Neuroscience, University of Montana, 32 Campus Drive, Missoula, MT 59812, USA
| | - Jesse C Hay
- Division of Biological Sciences & Center for Structural & Functional Neuroscience, University of Montana, 32 Campus Drive, Missoula, MT 59812, USA
| |
Collapse
|
15
|
Caffeine and MDMA (Ecstasy) Exacerbate ER Stress Triggered by Hyperthermia. Int J Mol Sci 2022; 23:ijms23041974. [PMID: 35216090 PMCID: PMC8880705 DOI: 10.3390/ijms23041974] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Revised: 02/03/2022] [Accepted: 02/07/2022] [Indexed: 02/04/2023] Open
Abstract
Drugs of abuse can cause local and systemic hyperthermia, a known trigger of endoplasmic reticulum (ER) stress and the unfolded protein response (UPR). Another trigger of ER stress and UPR is ER calcium depletion, which causes ER exodosis, the secretion of ER-resident proteins. In rodent models, club drugs such as 3,4-methylenedioxymethamphetamine (MDMA, ‘ecstasy’) can create hyperthermic conditions in the brain and cause toxicity that is affected by the environmental temperature and the presence of other drugs, such as caffeine. In human studies, MDMA stimulated an acute, dose-dependent increase in core body temperature, but an examination of caffeine and MDMA in combination remains a topic for clinical research. Here we examine the secretion of ER-resident proteins and activation of the UPR under combined exposure to MDMA and caffeine in a cellular model of hyperthermia. We show that hyperthermia triggers the secretion of normally ER-resident proteins, and that this aberrant protein secretion is potentiated by the presence of MDMA, caffeine, or a combination of the two drugs. Hyperthermia activates the UPR but the addition of MDMA or caffeine does not alter the canonical UPR gene expression despite the drug effects on ER exodosis of UPR-related proteins. One exception was increased BiP/GRP78 mRNA levels in MDMA-treated cells exposed to hyperthermia. These findings suggest that club drug use under hyperthermic conditions exacerbates disruption of ER proteostasis, contributing to cellular toxicity.
Collapse
|
16
|
Kalwat MA, Scheuner D, Rodrigues-dos-Santos K, Eizirik DL, Cobb MH. The Pancreatic ß-cell Response to Secretory Demands and Adaption to Stress. Endocrinology 2021; 162:bqab173. [PMID: 34407177 PMCID: PMC8459449 DOI: 10.1210/endocr/bqab173] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/25/2021] [Indexed: 02/06/2023]
Abstract
Pancreatic β cells dedicate much of their protein translation capacity to producing insulin to maintain glucose homeostasis. In response to increased secretory demand, β cells can compensate by increasing insulin production capability even in the face of protracted peripheral insulin resistance. The ability to amplify insulin secretion in response to hyperglycemia is a critical facet of β-cell function, and the exact mechanisms by which this occurs have been studied for decades. To adapt to the constant and fast-changing demands for insulin production, β cells use the unfolded protein response of the endoplasmic reticulum. Failure of these compensatory mechanisms contributes to both type 1 and 2 diabetes. Additionally, studies in which β cells are "rested" by reducing endogenous insulin demand have shown promise as a therapeutic strategy that could be applied more broadly. Here, we review recent findings in β cells pertaining to the metabolic amplifying pathway, the unfolded protein response, and potential advances in therapeutics based on β-cell rest.
Collapse
Affiliation(s)
- Michael A Kalwat
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
| | - Donalyn Scheuner
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
| | | | - Decio L Eizirik
- Indiana Biosciences Research Institute, Indianapolis, IN 46202, USA
- ULB Center for Diabetes Research, Medical Faculty, Université Libre de Bruxelles, Brussels, Belgium
| | - Melanie H Cobb
- Department of Pharmacology, University of Texas Southwestern Medical Center, Dallas, TX, 75390, USA
| |
Collapse
|
17
|
Jayanthi S, Daiwile AP, Cadet JL. Neurotoxicity of methamphetamine: Main effects and mechanisms. Exp Neurol 2021; 344:113795. [PMID: 34186102 PMCID: PMC8338805 DOI: 10.1016/j.expneurol.2021.113795] [Citation(s) in RCA: 106] [Impact Index Per Article: 26.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2021] [Revised: 06/03/2021] [Accepted: 06/24/2021] [Indexed: 12/12/2022]
Abstract
Methamphetamine (METH) is an illicit psychostimulant that is abused throughout the world. METH addiction is also a major public health concern and the abuse of large doses of the drug is often associated with serious neuropsychiatric consequences that may include agitation, anxiety, hallucinations, paranoia, and psychosis. Some human methamphetamine users can also suffer from attention, memory, and executive deficits. METH-associated neurological and psychiatric complications might be related, in part, to METH-induced neurotoxic effects. Those include altered dopaminergic and serotonergic functions, neuronal apoptosis, astrocytosis, and microgliosis. Here we have endeavored to discuss some of the main effects of the drug and have presented the evidence supporting certain of the molecular and cellular bases of METH neurotoxicity. The accumulated evidence suggests the involvement of transcription factors, activation of dealth pathways that emanate from mitochondria and endoplasmic reticulum (ER), and a role for neuroinflammatory mechanisms. Understanding the molecular processes involved in METH induced neurotoxicity should help in developing better therapeutic approaches that might also serve to attenuate or block the biological consequences of use of large doses of the drug by some humans who meet criteria for METH use disorder.
Collapse
Affiliation(s)
- Subramaniam Jayanthi
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program, Baltimore, MD 21224, United States of America
| | - Atul P Daiwile
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program, Baltimore, MD 21224, United States of America
| | - Jean Lud Cadet
- Molecular Neuropsychiatry Research Branch, NIDA Intramural Research Program, Baltimore, MD 21224, United States of America.
| |
Collapse
|
18
|
Meganathan K, Prakasam R, Baldridge D, Gontarz P, Zhang B, Urano F, Bonni A, Maloney SE, Turner TN, Huettner JE, Constantino JN, Kroll KL. Altered neuronal physiology, development, and function associated with a common chromosome 15 duplication involving CHRNA7. BMC Biol 2021; 19:147. [PMID: 34320968 PMCID: PMC8317352 DOI: 10.1186/s12915-021-01080-7] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2021] [Accepted: 06/30/2021] [Indexed: 01/05/2023] Open
Abstract
BACKGROUND Copy number variants (CNVs) linked to genes involved in nervous system development or function are often associated with neuropsychiatric disease. While CNVs involving deletions generally cause severe and highly penetrant patient phenotypes, CNVs leading to duplications tend instead to exhibit widely variable and less penetrant phenotypic expressivity among affected individuals. CNVs located on chromosome 15q13.3 affecting the alpha-7 nicotinic acetylcholine receptor subunit (CHRNA7) gene contribute to multiple neuropsychiatric disorders with highly variable penetrance. However, the basis of such differential penetrance remains uncharacterized. Here, we generated induced pluripotent stem cell (iPSC) models from first-degree relatives with a 15q13.3 duplication and analyzed their cellular phenotypes to uncover a basis for the dissimilar phenotypic expressivity. RESULTS The first-degree relatives studied included a boy with autism and emotional dysregulation (the affected proband-AP) and his clinically unaffected mother (UM), with comparison to unrelated control models lacking this duplication. Potential contributors to neuropsychiatric impairment were modeled in iPSC-derived cortical excitatory and inhibitory neurons. The AP-derived model uniquely exhibited disruptions of cellular physiology and neurodevelopment not observed in either the UM or unrelated controls. These included enhanced neural progenitor proliferation but impaired neuronal differentiation, maturation, and migration, and increased endoplasmic reticulum (ER) stress. Both the neuronal migration deficit and elevated ER stress could be selectively rescued by different pharmacologic agents. Neuronal gene expression was also dysregulated in the AP, including reduced expression of genes related to behavior, psychological disorders, neuritogenesis, neuronal migration, and Wnt, axonal guidance, and GABA receptor signaling. The UM model instead exhibited upregulated expression of genes in many of these same pathways, suggesting that molecular compensation could have contributed to the lack of neurodevelopmental phenotypes in this model. However, both AP- and UM-derived neurons exhibited shared alterations of neuronal function, including increased action potential firing and elevated cholinergic activity, consistent with increased homomeric CHRNA7 channel activity. CONCLUSIONS These data define both diagnosis-associated cellular phenotypes and shared functional anomalies related to CHRNA7 duplication that may contribute to variable phenotypic penetrance in individuals with 15q13.3 duplication. The capacity for pharmacological agents to rescue some neurodevelopmental anomalies associated with diagnosis suggests avenues for intervention for carriers of this duplication and other CNVs that cause related disorders.
Collapse
Affiliation(s)
- Kesavan Meganathan
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Avenue, Campus, Box 8103, St. Louis, MO 63110 USA
| | - Ramachandran Prakasam
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Avenue, Campus, Box 8103, St. Louis, MO 63110 USA
| | - Dustin Baldridge
- Department of Pediatrics, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Paul Gontarz
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Avenue, Campus, Box 8103, St. Louis, MO 63110 USA
| | - Bo Zhang
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Avenue, Campus, Box 8103, St. Louis, MO 63110 USA
| | - Fumihiko Urano
- Department of Medicine, Division of Endocrinology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Azad Bonni
- Department of Neuroscience, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Susan E. Maloney
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Tychele N. Turner
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - James E. Huettner
- Department of Cell Biology and Physiology, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - John N. Constantino
- Department of Psychiatry, Washington University School of Medicine, St. Louis, MO 63110 USA
| | - Kristen L. Kroll
- Department of Developmental Biology, Washington University School of Medicine, 660 S. Euclid Avenue, Campus, Box 8103, St. Louis, MO 63110 USA
| |
Collapse
|
19
|
Trychta KA, Xie B, Verma RK, Xu M, Shi L, Harvey BK. Computational Modeling of C-Terminal Tails to Predict the Calcium-Dependent Secretion of Endoplasmic Reticulum Resident Proteins. Front Chem 2021; 9:689608. [PMID: 34268295 PMCID: PMC8276033 DOI: 10.3389/fchem.2021.689608] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2021] [Accepted: 06/03/2021] [Indexed: 12/29/2022] Open
Abstract
The lumen of the endoplasmic reticulum (ER) has resident proteins that are critical to perform the various tasks of the ER such as protein maturation and lipid metabolism. These ER resident proteins typically have a carboxy-terminal ER retention/retrieval sequence (ERS). The canonical ERS that promotes ER retrieval is Lys-Asp-Glu-Leu (KDEL) and when an ER resident protein moves from the ER to the Golgi, KDEL receptors (KDELRs) in the Golgi recognize the ERS and return the protein to the ER lumen. Depletion of ER calcium leads to the mass departure of ER resident proteins in a process termed exodosis, which is regulated by KDELRs. Here, by combining computational prediction with machine learning-based models and experimental validation, we identify carboxy tail sequences of ER resident proteins divergent from the canonical “KDEL” ERS. Using molecular modeling and simulations, we demonstrated that two representative non-canonical ERS can stably bind to the KDELR. Collectively, we developed a method to predict whether a carboxy-terminal sequence acts as a putative ERS that would undergo secretion in response to ER calcium depletion and interacts with the KDELRs. The interaction between the ERS and the KDELR extends beyond the final four carboxy terminal residues of the ERS. Identification of proteins that undergo exodosis will further our understanding of changes in ER proteostasis under physiological and pathological conditions where ER calcium is depleted.
Collapse
Affiliation(s)
- Kathleen A Trychta
- Molecular Mechanisms of Cellular Stress and Inflammation Unit, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Bing Xie
- Computational Chemistry and Molecular Biophysics Section, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Ravi Kumar Verma
- Computational Chemistry and Molecular Biophysics Section, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Min Xu
- Computational Chemistry and Molecular Biophysics Section, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Lei Shi
- Computational Chemistry and Molecular Biophysics Section, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Brandon K Harvey
- Molecular Mechanisms of Cellular Stress and Inflammation Unit, National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| |
Collapse
|
20
|
The Function of KDEL Receptors as UPR Genes in Disease. Int J Mol Sci 2021; 22:ijms22115436. [PMID: 34063979 PMCID: PMC8196686 DOI: 10.3390/ijms22115436] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2021] [Revised: 05/17/2021] [Accepted: 05/17/2021] [Indexed: 12/15/2022] Open
Abstract
The KDEL receptor retrieval pathway is essential for maintaining resident proteins in the endoplasmic reticulum (ER) lumen. ER resident proteins serve a variety of functions, including protein folding and maturation. Perturbations to the lumenal ER microenvironment, such as calcium depletion, can cause protein misfolding and activation of the unfolded protein response (UPR). Additionally, ER resident proteins are secreted from the cell by overwhelming the KDEL receptor retrieval pathway. Recent data show that KDEL receptors are also activated during the UPR through the IRE1/XBP1 signaling pathway as an adaptive response to cellular stress set forth to reduce the loss of ER resident proteins. This review will discuss the emerging connection between UPR activation and KDEL receptors as it pertains to ER proteostasis and disease states.
Collapse
|
21
|
Henderson MJ, Trychta KA, Yang SM, Bäck S, Yasgar A, Wires ES, Danchik C, Yan X, Yano H, Shi L, Wu KJ, Wang AQ, Tao D, Zahoránszky-Kőhalmi G, Hu X, Xu X, Maloney D, Zakharov AV, Rai G, Urano F, Airavaara M, Gavrilova O, Jadhav A, Wang Y, Simeonov A, Harvey BK. A target-agnostic screen identifies approved drugs to stabilize the endoplasmic reticulum-resident proteome. Cell Rep 2021; 35:109040. [PMID: 33910017 DOI: 10.1016/j.celrep.2021.109040] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2020] [Revised: 02/12/2021] [Accepted: 04/06/2021] [Indexed: 12/24/2022] Open
Abstract
Endoplasmic reticulum (ER) dysregulation is associated with pathologies including neurodegenerative, muscular, and diabetic conditions. Depletion of ER calcium can lead to the loss of resident proteins in a process termed exodosis. To identify compounds that attenuate the redistribution of ER proteins under pathological conditions, we performed a quantitative high-throughput screen using the Gaussia luciferase (GLuc)-secreted ER calcium modulated protein (SERCaMP) assay, which monitors secretion of ER-resident proteins triggered by calcium depletion. We identify several clinically used drugs, including bromocriptine, and further characterize them using assays to measure effects on ER calcium, ER stress, and ER exodosis. Bromocriptine elicits protective effects in cell-based models of exodosis as well as in vivo models of stroke and diabetes. Bromocriptine analogs with reduced dopamine receptor activity retain similar efficacy in stabilizing the ER proteome, indicating a non-canonical mechanism of action. This study describes a strategic approach to identify small-molecule drugs capable of improving ER proteostasis in human disease conditions.
Collapse
Affiliation(s)
- Mark J Henderson
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA.
| | - Kathleen A Trychta
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Shyh-Ming Yang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Susanne Bäck
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Adam Yasgar
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Emily S Wires
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Carina Danchik
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Xiaokang Yan
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Hideaki Yano
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Lei Shi
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA
| | - Kuo-Jen Wu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Amy Q Wang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Dingyin Tao
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Gergely Zahoránszky-Kőhalmi
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Xin Hu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Xin Xu
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - David Maloney
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Alexey V Zakharov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Ganesha Rai
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Fumihiko Urano
- Department of Medicine, Division of Endocrinology, Metabolism, and Lipid Research, Washington University in St. Louis, St. Louis, MO 63110, USA
| | - Mikko Airavaara
- Neuroscience Center, HiLIFE & Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Oksana Gavrilova
- National Institute of Diabetes and Digestive and Kidney Diseases, Bethesda, MD 20892, USA
| | - Ajit Jadhav
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan 35053, Taiwan
| | - Anton Simeonov
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850, USA
| | - Brandon K Harvey
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD 21224, USA.
| |
Collapse
|
22
|
Jӓntti M, Harvey BK. Trophic activities of endoplasmic reticulum proteins CDNF and MANF. Cell Tissue Res 2020; 382:83-100. [PMID: 32845431 DOI: 10.1007/s00441-020-03263-0] [Citation(s) in RCA: 35] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2020] [Accepted: 07/20/2020] [Indexed: 12/13/2022]
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) and cerebral dopamine neurotrophic factor (CDNF) are endoplasmic reticulum (ER) luminal proteins that confer trophic activities in a wide range of tissues under diverse pathological conditions. Despite initially being classified as neurotrophic factors, neither protein structurally nor functionally resembles bona fide neurotrophic factors. Their highly homologous structures comprise a unique globular, saposin-like domain within the N-terminus joined by a flexible linker to a C-terminus containing a SAP-like domain, CXXC motif and an ER retention sequence. Neurotrophic factors exert effects by binding to cognate receptors in the plasma membrane; however, no cell surface receptors have been identified for MANF and CDNF. Both can act as unfolded protein response (UPR) genes that modulate the UPR and inflammatory processes. The trophic activity of MANF and CDNF extends beyond the central nervous system with MANF being crucial for the development of pancreatic β cells and both have trophic effects in a variety of diseases related to the liver, heart, skeletal tissue, kidney and peripheral nervous system. In this article, the unique features of MANF and CDNF, such as their structure and mechanisms of action related to ER stress and inflammation, will be reviewed. Recently identified interactions with lipids and membrane trafficking will also be described. Lastly, their function and therapeutic potential in different diseases including a recent clinical trial using CDNF to treat Parkinson's disease will be discussed. Collectively, this review will highlight MANF and CDNF as broad-acting trophic factors that regulate functions of the endoplasmic reticulum.
Collapse
Affiliation(s)
- Maria Jӓntti
- Molecular Mechanisms of Cellular Stress and Inflammation Lab, Intramural Research Program, National Institute on Drug Abuse, Suite 200, 251 Bayview Blvd, Baltimore, MD, 21224, USA
| | - Brandon K Harvey
- Molecular Mechanisms of Cellular Stress and Inflammation Lab, Intramural Research Program, National Institute on Drug Abuse, Suite 200, 251 Bayview Blvd, Baltimore, MD, 21224, USA.
| |
Collapse
|
23
|
Lawal TA, Wires ES, Terry NL, Dowling JJ, Todd JJ. Preclinical model systems of ryanodine receptor 1-related myopathies and malignant hyperthermia: a comprehensive scoping review of works published 1990-2019. Orphanet J Rare Dis 2020; 15:113. [PMID: 32381029 PMCID: PMC7204063 DOI: 10.1186/s13023-020-01384-x] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2019] [Accepted: 04/14/2020] [Indexed: 02/07/2023] Open
Abstract
BACKGROUND Pathogenic variations in the gene encoding the skeletal muscle ryanodine receptor (RyR1) are associated with malignant hyperthermia (MH) susceptibility, a life-threatening hypermetabolic condition and RYR1-related myopathies (RYR1-RM), a spectrum of rare neuromuscular disorders. In RYR1-RM, intracellular calcium dysregulation, post-translational modifications, and decreased protein expression lead to a heterogenous clinical presentation including proximal muscle weakness, contractures, scoliosis, respiratory insufficiency, and ophthalmoplegia. Preclinical model systems of RYR1-RM and MH have been developed to better understand underlying pathomechanisms and test potential therapeutics. METHODS We conducted a comprehensive scoping review of scientific literature pertaining to RYR1-RM and MH preclinical model systems in accordance with the PRISMA Scoping Reviews Checklist and the framework proposed by Arksey and O'Malley. Two major electronic databases (PubMed and EMBASE) were searched without language restriction for articles and abstracts published between January 1, 1990 and July 3, 2019. RESULTS Our search yielded 5049 publications from which 262 were included in this review. A majority of variants tested in RYR1 preclinical models were localized to established MH/central core disease (MH/CCD) hot spots. A total of 250 unique RYR1 variations were reported in human/rodent/porcine models with 95% being missense substitutions. The most frequently reported RYR1 variant was R614C/R615C (human/porcine total n = 39), followed by Y523S/Y524S (rabbit/mouse total n = 30), I4898T/I4897T/I4895T (human/rabbit/mouse total n = 20), and R163C/R165C (human/mouse total n = 18). The dyspedic mouse was utilized by 47% of publications in the rodent category and its RyR1-null (1B5) myotubes were transfected in 23% of publications in the cellular model category. In studies of transfected HEK-293 cells, 57% of RYR1 variations affected the RyR1 channel and activation core domain. A total of 15 RYR1 mutant mouse strains were identified of which ten were heterozygous, three were compound heterozygous, and a further two were knockout. Porcine, avian, zebrafish, C. elegans, canine, equine, and drosophila model systems were also reported. CONCLUSIONS Over the past 30 years, there were 262 publications on MH and RYR1-RM preclinical model systems featuring more than 200 unique RYR1 variations tested in a broad range of species. Findings from these studies have set the foundation for therapeutic development for MH and RYR1-RM.
Collapse
Affiliation(s)
- Tokunbor A Lawal
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, 20892, USA
| | - Emily S Wires
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, USA
| | - Nancy L Terry
- National Institutes of Health Library, National Institutes of Health, Bethesda, MD, USA
| | - James J Dowling
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, Ontario, Canada
| | - Joshua J Todd
- National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, 20892, USA.
| |
Collapse
|
24
|
Volpatti JR, Endo Y, Knox J, Groom L, Brennan S, Noche R, Zuercher WJ, Roy P, Dirksen RT, Dowling JJ. Identification of drug modifiers for RYR1-related myopathy using a multi-species discovery pipeline. eLife 2020; 9:52946. [PMID: 32223895 PMCID: PMC7202896 DOI: 10.7554/elife.52946] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2019] [Accepted: 03/29/2020] [Indexed: 02/06/2023] Open
Abstract
Ryanodine receptor type I-related myopathies (RYR1-RMs) are a common group of childhood muscle diseases associated with severe disabilities and early mortality for which there are no available treatments. The goal of this study is to identify new therapeutic targets for RYR1-RMs. To accomplish this, we developed a discovery pipeline using nematode, zebrafish, and mammalian cell models. We first performed large-scale drug screens in C. elegans which uncovered 74 hits. Targeted testing in zebrafish yielded positive results for two p38 inhibitors. Using mouse myotubes, we found that either pharmacological inhibition or siRNA silencing of p38 impaired caffeine-induced Ca2+ release from wild type cells while promoting intracellular Ca2+ release in Ryr1 knockout cells. Lastly, we demonstrated that p38 inhibition blunts the aberrant temperature-dependent increase in resting Ca2+ in myotubes from an RYR1-RM mouse model. This unique platform for RYR1-RM therapy development is potentially applicable to a broad range of neuromuscular disorders. Muscle cells have storage compartments stuffed full of calcium, which they release to trigger a contraction. This process depends on a channel-shaped protein called the ryanodine receptor, or RYR1 for short. When RYR1 is activated, it releases calcium from storage, which floods the muscle cell. Mutations in the gene that codes for RYR1 in humans cause a group of rare diseases called RYR1-related myopathies. The mutations change calcium release in muscle cells, which can make movement difficult, and make it hard for people to breathe. At the moment, RYR1 myopathies have no treatment. It is possible that repurposing existing drugs could benefit people with RYR1-related myopathies, but trialing treatments takes time. The fastest and cheapest way to test whether compounds might be effective is to try them on very simple animals, like nematode worms. But even though worms and humans share certain genes, treatments that work for worms do not always work for humans. Luckily, it is sometimes possible to test whether compounds might be effective by trying them out on complex mammals, like mice. Unfortunately, these experiments are slow and expensive. A compromise involves testing on animals such as zebrafish. So far, none of these methods has been successful in discovering treatments for RYR1-related myopathies. To maximize the strengths of each animal model, Volpatti et al. combined them, developing a fast and powerful way to test new drugs. The first step is an automated screening process that trials thousands of chemicals on nematode worms. This takes just two weeks. The second step is to group the best treatments according to their chemical similarities and test them again in zebrafish. This takes a month. The third and final stage is to test promising chemicals from the zebrafish in mouse muscle cells. Of the thousands of compounds tested here, one group of chemicals stood out – treatments that block the activity of a protein called p38. Volpatti et al. found that blocking the p38 protein, either with drugs or by inactivating the gene that codes for it, changed muscle calcium release. This suggests p38 blockers may have potential as a treatment for RYR1-related myopathies in mammals. Using three types of animal to test new drugs maximizes the benefits of each model. This type of pipeline could identify new treatments, not just for RYR1-related myopathies, but for other diseases that involve genes or proteins that are similar across species. For RYR1-related myopathies specifically, the next step is to test p38 blocking treatments in mice. This could reveal whether the treatments have the potential to improve symptoms.
Collapse
Affiliation(s)
- Jonathan R Volpatti
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Yukari Endo
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, Canada
| | - Jessica Knox
- Department of Molecular Genetics, University of Toronto, Toronto, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Linda Groom
- Department of Pharmacology, University of Rochester, Rochester, United States
| | - Stephanie Brennan
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| | - Ramil Noche
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, Canada
| | - William J Zuercher
- UNC Eshelman School of Pharmacy, SGC Center for Chemical Biology, University of North Carolina, Chapel Hill, United States
| | - Peter Roy
- Department of Molecular Genetics, University of Toronto, Toronto, Canada.,Department of Pharmacology and Toxicology, University of Toronto, Toronto, Canada
| | - Robert T Dirksen
- Department of Pharmacology, University of Rochester, Rochester, United States
| | - James J Dowling
- Program for Genetics and Genome Biology, Hospital for Sick Children, Toronto, Canada.,Department of Molecular Genetics, University of Toronto, Toronto, Canada
| |
Collapse
|
25
|
Yu SJ, Wu KJ, Bae E, Wang YS, Chiang CW, Kuo LW, Harvey BK, Greig NH, Wang Y. Post-treatment with Posiphen Reduces Endoplasmic Reticulum Stress and Neurodegeneration in Stroke Brain. iScience 2020; 23:100866. [PMID: 32058974 PMCID: PMC7013187 DOI: 10.1016/j.isci.2020.100866] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2019] [Revised: 12/18/2019] [Accepted: 01/21/2020] [Indexed: 12/26/2022] Open
Abstract
Acetylcholinesterase (AChE) inhibitors have protective and anti-inflammatory actions against brain injury, mediated by nicotinic α7 cholinergic receptor activation. The use of AChE inhibitors in patients is limited by systemic cholinergic side effects. Posiphen, a stereoisomer of the AChE inhibitor Phenserine, lacks AChE inhibitor activity. The purpose of this study is to determine the protective effect of Posiphen in cellular and animal models of stroke. Both Posiphen and Phenserine reduced glutamate-mediated neuronal loss in co-cultures of primary cortical cells and microglia. Phenserine-, but not Posiphen-, mediated neuroprotection was diminished by the nicotinic α7 receptor antagonist methyllycaconitine. Posiphen antagonized NMDA-mediated Ca++ influx, thapsigargin-mediated neuronal loss and ER stress in cultured cells. Early post-treatment with Posiphen reduced ER stress signals, IBA1 immunoreactivity, TUNEL and infarction in the ischemic cortex, as well as neurological deficits in stroke rats. These findings indicate that Posiphen is neuroprotective against stroke through regulating Ca++i and ER stress. Posiphen induces protection in cell culture through noncholinergic mechanism Posiphen attenuates glutamate-mediated Ca++i and ER stress in neuronal culture Posiphen mitigates ER stress in stroke brain Posiphen reduces neurodegeneration in stroke rats
Collapse
Affiliation(s)
- Seong-Jin Yu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Kuo-Jen Wu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Eunkyung Bae
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Yu-Syuan Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Chia-Wen Chiang
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Miaoli, Taiwan
| | - Li-Wei Kuo
- Institute of Biomedical Engineering and Nanomedicine, National Health Research Institutes, Zhunan, Miaoli, Taiwan
| | | | - Nigel H Greig
- Translational Gerontology Branch, Intramural Research Program, National Institute of Aging, NIH, Baltimore, MD, USA
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan.
| |
Collapse
|
26
|
KDEL Receptors Are Differentially Regulated to Maintain the ER Proteome under Calcium Deficiency. Cell Rep 2019; 25:1829-1840.e6. [PMID: 30428351 DOI: 10.1016/j.celrep.2018.10.055] [Citation(s) in RCA: 85] [Impact Index Per Article: 14.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/10/2018] [Revised: 09/17/2018] [Accepted: 10/12/2018] [Indexed: 12/21/2022] Open
Abstract
Retention of critical endoplasmic reticulum (ER) luminal proteins needed to carry out diverse functions (e.g., protein synthesis and folding, lipid metabolism) is mediated through a carboxy-terminal ER retention sequence (ERS) and its interaction with KDEL receptors. Here, we demonstrate that depleting ER calcium causes mass departure of ERS-containing proteins from cells by overwhelming KDEL receptors. In addition, we provide evidence that KDELR2 and KDELR3, but not KDELR1, are unfolded protein response (UPR) genes upregulated as an adaptive response to counteract the loss of ERS-containing proteins, suggesting previously unknown isoform-specific functions of the KDEL receptors. Overall, our findings establish that decreases in ER calcium change the composition of the ER luminal proteome and secretome, which can impact cellular functions and cell viability. The redistribution of the ER proteome from inside the cell to the outside has implications for dissecting the complex relationship of ER homeostasis with diverse disease pathologies.
Collapse
|
27
|
Park SJ, Kim Y, Yang SM, Henderson MJ, Yang W, Lindahl M, Urano F, Chen YM. Discovery of endoplasmic reticulum calcium stabilizers to rescue ER-stressed podocytes in nephrotic syndrome. Proc Natl Acad Sci U S A 2019; 116:14154-14163. [PMID: 31235574 PMCID: PMC6628787 DOI: 10.1073/pnas.1813580116] [Citation(s) in RCA: 46] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023] Open
Abstract
Emerging evidence has established primary nephrotic syndrome (NS), including focal segmental glomerulosclerosis (FSGS), as a primary podocytopathy. Despite the underlying importance of podocyte endoplasmic reticulum (ER) stress in the pathogenesis of NS, no treatment currently targets the podocyte ER. In our monogenic podocyte ER stress-induced NS/FSGS mouse model, the podocyte type 2 ryanodine receptor (RyR2)/calcium release channel on the ER was phosphorylated, resulting in ER calcium leak and cytosolic calcium elevation. The altered intracellular calcium homeostasis led to activation of calcium-dependent cytosolic protease calpain 2 and cleavage of its important downstream substrates, including the apoptotic molecule procaspase 12 and podocyte cytoskeletal protein talin 1. Importantly, a chemical compound, K201, can block RyR2-Ser2808 phosphorylation-mediated ER calcium depletion and podocyte injury in ER-stressed podocytes, as well as inhibit albuminuria in our NS model. In addition, we discovered that mesencephalic astrocyte-derived neurotrophic factor (MANF) can revert defective RyR2-induced ER calcium leak, a bioactivity for this ER stress-responsive protein. Thus, podocyte RyR2 remodeling contributes to ER stress-induced podocyte injury. K201 and MANF could be promising therapies for the treatment of podocyte ER stress-induced NS/FSGS.
Collapse
Affiliation(s)
- Sun-Ji Park
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Yeawon Kim
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Shyh-Ming Yang
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Mark J Henderson
- National Center for Advancing Translational Sciences, National Institutes of Health, Rockville, MD 20850
| | - Wei Yang
- Department of Genetics, Washington University School of Medicine, St. Louis, MO 63110
| | - Maria Lindahl
- Institute of Biotechnology, University of Helsinki, Helsinki, Finland 00014
| | - Fumihiko Urano
- Division of Endocrinology, Metabolism, and Lipid Research, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110
| | - Ying Maggie Chen
- Division of Nephrology, Department of Medicine, Washington University School of Medicine, St. Louis, MO 63110;
| |
Collapse
|
28
|
Neuronal Activation Stimulates Cytomegalovirus Promoter-Driven Transgene Expression. MOLECULAR THERAPY-METHODS & CLINICAL DEVELOPMENT 2019; 14:180-188. [PMID: 31380464 PMCID: PMC6661544 DOI: 10.1016/j.omtm.2019.06.006] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/20/2019] [Accepted: 06/23/2019] [Indexed: 01/23/2023]
Abstract
The cytomegalovirus (CMV) immediate early promoter has been extensively developed and exploited for transgene expression in vitro and in vivo, including human clinical trials. The CMV promoter has long been considered a stable, constitutive, and ubiquitous promoter for transgene expression. Using two different CMV-based promoters, we found an increase in CMV-driven transgene expression in the rodent brain and in primary neuronal cultures in response to methamphetamine, glutamate, kainic acid, and activation of G protein-coupled receptor signaling using designer receptors exclusively activated by designer drugs (DREADDs). In contrast, promoters derived from human synapsin 1 (hSYN1) gene or elongation factor 1α (EF1α) did not exhibit altered transgene expression in response to the same neuronal stimulations. Overall, our results suggest that the long-standing assertion that the CMV promoter confers constitutive expression in neurons should be reevaluated, and future studies should empirically determine the activity of the CMV promoter in a given application.
Collapse
|
29
|
Pifithrin-Alpha Reduces Methamphetamine Neurotoxicity in Cultured Dopaminergic Neurons. Neurotox Res 2019; 36:347-356. [DOI: 10.1007/s12640-019-00050-w] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2019] [Revised: 03/26/2019] [Accepted: 04/16/2019] [Indexed: 12/28/2022]
|
30
|
Albert K, Voutilainen MH, Domanskyi A, Piepponen TP, Ahola S, Tuominen RK, Richie C, Harvey BK, Airavaara M. Downregulation of tyrosine hydroxylase phenotype after AAV injection above substantia nigra: Caution in experimental models of Parkinson's disease. J Neurosci Res 2019; 97:346-361. [PMID: 30548446 PMCID: PMC11863348 DOI: 10.1002/jnr.24363] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2018] [Revised: 11/19/2018] [Accepted: 11/19/2018] [Indexed: 12/14/2022]
Abstract
Adeno-associated virus (AAV) vector-mediated delivery of human α-synuclein (α-syn) gene in rat substantia nigra (SN) results in increased expression of α-syn protein in the SN and striatum which can progressively degenerate dopaminergic neurons. Therefore, this model is thought to recapitulate the neurodegeneration in Parkinson's disease. Here, using AAV to deliver α-syn above the SN in male and female rats resulted in clear expression of human α-syn in the SN and striatum. The protein was associated with moderate behavioral deficits and some loss of tyrosine hydroxylase (TH) in the nigrostriatal areas. However, the immunohistochemistry results were highly variable and showed little to no correlation with behavior and the amount of α-syn present. Expression of green fluorescent protein (GFP) was used as a control to monitor gene delivery and expression efficacy. AAV-GFP resulted in a similar or greater TH loss compared to AAV-α-syn and therefore an additional vector that does not express a protein was tested. Vectors with double-floxed inverse open reading frame (DIO ORF) encoding fluorescent proteins that generate RNA that is not translated also resulted in TH downregulation in the SN but showed no significant behavioral deficits. These results demonstrate that although expression of wild-type human α-syn can cause neurodegeneration, the variability and lack of correlation with outcome measures are drawbacks with the model. Furthermore, design and control selection should be considered carefully because of conflicting conclusions due to AAV downregulation of TH, and we recommend caution with having highly regulated TH as the only marker for the dopamine system.
Collapse
Affiliation(s)
- Katrina Albert
- Institute of Biotechnology, Program of Developmental Biology, University of Helsinki, Helsinki, Finland
| | - Merja H. Voutilainen
- Institute of Biotechnology, Program of Developmental Biology, University of Helsinki, Helsinki, Finland
| | - Andrii Domanskyi
- Institute of Biotechnology, Program of Developmental Biology, University of Helsinki, Helsinki, Finland
| | - T. Petteri Piepponen
- Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
| | - Sari Ahola
- Institute of Biotechnology, Program of Developmental Biology, University of Helsinki, Helsinki, Finland
| | - Raimo K. Tuominen
- Division of Pharmacology and Pharmacotherapy, University of Helsinki, Helsinki, Finland
| | - Christopher Richie
- Intramural Research Program, National Institute on Drug Abuse, NIH, Baltimore, MD, USA
| | - Brandon K. Harvey
- Intramural Research Program, National Institute on Drug Abuse, NIH, Baltimore, MD, USA
| | - Mikko Airavaara
- Institute of Biotechnology, Program of Developmental Biology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
31
|
Kearney SE, Zahoránszky-Kőhalmi G, Brimacombe KR, Henderson MJ, Lynch C, Zhao T, Wan K, Itkin Z, Dillon C, Shen M, Cheff D, Lee T, Bougie D, Cheng K, Coussens N, Dorjsuren D, Eastman R, Huang R, Iannotti M, Karavadhi S, Klumpp-Thomas C, Roth J, Sakamuru S, Sun W, Titus S, Yasgar A, Zhang YQ, Zhao J, Andrade R, Brown MK, Burns N, Cha JK, Mevers E, Clardy J, Clement J, Crooks P, Cuny G, Ganor J, Moreno J, Morrill L, Picazo E, Susick R, Garg N, Goess B, Grossman R, Hughes C, Johnston J, Joullie M, Kinghorn AD, Kingston D, Krische M, Kwon O, Maimone T, Majumdar S, Maloney K, Mohamed E, Murphy B, Nagorny P, Olson D, Overman L, Brown L, Snyder J, Porco J, Rivas F, Ross S, Sarpong R, Sharma I, Shaw J, Xu Z, Shen B, Shi W, Stephenson C, Verano A, Tan D, Tang Y, Taylor R, Thomson R, Vosburg D, Wu J, Wuest W, Zakarian A, Zhang Y, Ren T, Zuo Z, Inglese J, Michael S, Simeonov A, Zheng W, Shinn P, Jadhav A, Boxer M, Hall MD, Xia M, Guha R, Rohde JM. Canvass: A Crowd-Sourced, Natural-Product Screening Library for Exploring Biological Space. ACS CENTRAL SCIENCE 2018; 4:1727-1741. [PMID: 30648156 PMCID: PMC6311695 DOI: 10.1021/acscentsci.8b00747] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/12/2018] [Indexed: 05/20/2023]
Abstract
Natural products and their derivatives continue to be wellsprings of nascent therapeutic potential. However, many laboratories have limited resources for biological evaluation, leaving their previously isolated or synthesized compounds largely or completely untested. To address this issue, the Canvass library of natural products was assembled, in collaboration with academic and industry researchers, for quantitative high-throughput screening (qHTS) across a diverse set of cell-based and biochemical assays. Characterization of the library in terms of physicochemical properties, structural diversity, and similarity to compounds in publicly available libraries indicates that the Canvass library contains many structural elements in common with approved drugs. The assay data generated were analyzed using a variety of quality control metrics, and the resultant assay profiles were explored using statistical methods, such as clustering and compound promiscuity analyses. Individual compounds were then sorted by structural class and activity profiles. Differential behavior based on these classifications, as well as noteworthy activities, are outlined herein. One such highlight is the activity of (-)-2(S)-cathafoline, which was found to stabilize calcium levels in the endoplasmic reticulum. The workflow described here illustrates a pilot effort to broadly survey the biological potential of natural products by utilizing the power of automation and high-throughput screening.
Collapse
Affiliation(s)
- Sara E. Kearney
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Gergely Zahoránszky-Kőhalmi
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Kyle R. Brimacombe
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Mark J. Henderson
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Caitlin Lynch
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Tongan Zhao
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Kanny
K. Wan
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
- Department
of Chemistry, Harvey Mudd College, 301 Platt Boulevard, Claremont, California 91711, United States
| | - Zina Itkin
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Christopher Dillon
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Min Shen
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Dorian
M. Cheff
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Tobie
D. Lee
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Danielle Bougie
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Ken Cheng
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Nathan
P. Coussens
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Dorjbal Dorjsuren
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Richard
T. Eastman
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Ruili Huang
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Michael
J. Iannotti
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Surendra Karavadhi
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Carleen Klumpp-Thomas
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Jacob
S. Roth
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Srilatha Sakamuru
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Wei Sun
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Steven
A. Titus
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Adam Yasgar
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Ya-Qin Zhang
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Jinghua Zhao
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Rodrigo
B. Andrade
- Department
of Chemistry, Temple University, 1901 North 13th Street, Philadelphia, Pennsylvania 19122, United States
| | - M. Kevin Brown
- Department
of Chemistry, Indiana University, 800 East Kirkwood Avenue, Bloomington, Indiana 47405, United States
| | - Noah
Z. Burns
- Department
of Chemistry, Stanford University, 333 Campus Drive, Stanford, California 94305, United States
| | - Jin K. Cha
- Department
of Chemistry, Wayne State University, 5101 Cass Avenue, Detroit, Michigan 48202, United States
| | - Emily
E. Mevers
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Jon Clardy
- Department
of Biological Chemistry and Molecular Pharmacology, Harvard Medical School, 240 Longwood Avenue, Boston, Massachusetts 02115, United States
| | - Jason
A. Clement
- Natural
Products Discovery Institute, Baruch S. Blumberg Institute, 3805 Old Easton Road, Doylestown, Pennsylvania 18902, United States
| | - Peter
A. Crooks
- University
of Arkansas for Medical Sciences, 4301 West Markham Street 522, Little Rock, Arkansas 72205, United States
| | - Gregory
D. Cuny
- Department
of Pharmacological and Pharmaceutical Sciences, University of Houston, 4849 Calhoun Road, Houston, Texas 77204, United
States
| | - Jake Ganor
- Diamond
Age Corp., 344 East Louisiana
Street, McKinney, Texas 75069, United States
| | - Jesus Moreno
- Department
of Chemistry and Biochemistry, UCLA, 607 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Lucas
A. Morrill
- Department
of Chemistry and Biochemistry, UCLA, 607 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Elias Picazo
- Department
of Chemistry and Biochemistry, UCLA, 607 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Robert
B. Susick
- Department
of Chemistry and Biochemistry, UCLA, 607 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Neil
K. Garg
- Department
of Chemistry and Biochemistry, UCLA, 607 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Brian
C. Goess
- Department
of Chemistry, Furman University, 3300 Poinsett Highway, Greenville, South Carolina 29613, United States
| | - Robert
B. Grossman
- Department
of Chemistry, University of Kentucky, Lexington, Kentucky 40506, United States
| | - Chambers
C. Hughes
- Scripps
Institution of Oceanography, UCSD, 9500 Gilman Drive, La Jolla, California 92093, United States
| | - Jeffrey
N. Johnston
- Department
of Chemistry, Vanderbilt University, 7330 Stevenson Center, Nashville, Tennessee 37235, United States
| | - Madeleine
M. Joullie
- Department
of Chemistry, University of Pennsylvania, 231 South 34th Street, Philadelphia, Pennsylvania 19104, United States
| | - A. Douglas Kinghorn
- College
of Pharmacy, The Ohio State University, 500 West 12th Avenue, Columbus, Ohio 43210, United States
| | - David
G.I. Kingston
- Department
of Chemistry, Virginia Tech, 900 West Campus Drive, Blacksburg, Virginia 24061, United States
| | - Michael
J. Krische
- Chemistry
Department, The University of Texas at Austin, 105 East 24th Street STOP A5300, Austin, Texas 78712, United States
| | - Ohyun Kwon
- Department
of Chemistry and Biochemistry, UCLA, 607 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Thomas
J. Maimone
- Department
of Chemistry, University of California Berkeley, 826 Latimer Hall, Berkeley, California 94720, United States
| | - Susruta Majumdar
- Department
of Molecular Pharmacology and Neurology, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
- Center for
Clinical Pharmacology, St Louis College
of Pharmacy and Washington University School of Medicine, 2 Pharmacy Place, St. Louis, Missouri 63110, United States
| | - Katherine
N. Maloney
- Department
of Chemistry, Point Loma Nazarene University, 3900 Lomaland Drive, San Diego, California 92106, United States
| | - Enas Mohamed
- University
of Mississippi School of Pharmacy, 2500 North State Street, Jackson, Mississippi 39216, United States
| | - Brian
T. Murphy
- College
of Pharmacy, Department of Medicinal Chemistry and Pharmacognosy, University of Illinois at Chicago, 900 South Ashland Avenue, Chicago, Illinois 60607, United States
| | - Pavel Nagorny
- Department
of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| | - David
E. Olson
- Department
of Chemistry, University of California, Davis, One Shields Avenue, Davis, California 95616, United States
- School of
Medicine, Department of Biochemistry and Molecular Medicine, University of California, Davis, 2700 Stockton Boulevard, Suite 2102, Sacramento, California 95817, United States
- Center for
Neuroscience, University of California,
Davis, 1544 Newton Court, Davis, California 95618, United States
| | - Larry
E. Overman
- Department
of Chemistry, University of California,
Irvine, Irvine, California 92697, United States
| | - Lauren
E. Brown
- Department
of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
| | - John
K. Snyder
- Department
of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
| | - John
A. Porco
- Department
of Chemistry and Center for Molecular Discovery (BU-CMD), Boston University, 590 Commonwealth Avenue, Boston, Massachusetts 02215, United States
| | - Fatima Rivas
- Department
of Chemical Biology and Therapeutics, St.
Jude Children’s Research Hospital, 262 Danny Thomas Place, Memphis, Tennessee 38105, United States
| | - Samir
A. Ross
- University
of Mississippi School of Pharmacy, 2500 North State Street, Jackson, Mississippi 39216, United States
| | - Richmond Sarpong
- Department
of Chemistry, University of California Berkeley, 841-A Latimer Hall, Berkeley, California 94720, United States
| | - Indrajeet Sharma
- Department
of Chemistry and Biochemistry, and Institute of Natural Products and
Research Technologies, University of Oklahoma, 101 Stephenson Parkway, Norman, Oklahoma 73019, United States
| | - Jared
T. Shaw
- Department
of Chemistry, University of California, Davis, One Shields Avenue, Davis, California 95616, United States
| | - Zhengren Xu
- Department
of Chemistry, Florida Campus, The Scripps
Research Institute, 130
Scripps Way, Jupiter, Florida 33458, United States
| | - Ben Shen
- Department
of Chemistry, Florida Campus, The Scripps
Research Institute, 130
Scripps Way, Jupiter, Florida 33458, United States
| | - Wei Shi
- Department
of Chemistry and Biochemistry, University
of Arkansas, Fayetteville, Arkansas 72701, United States
| | - Corey
R.J. Stephenson
- Department
of Chemistry, University of Michigan, 930 North University Avenue, Ann Arbor, Michigan 48109, United States
| | - Alyssa
L. Verano
- Pharmacology
Graduate Program, Weill Cornell Graduate School of Medical Sciences, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
| | - Derek
S. Tan
- Pharmacology
Graduate Program, Weill Cornell Graduate School of Medical Sciences, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, New York, New York 10065, United States
- Chemical
Biology Program, Sloan Kettering Institute and Tri-Institutional Research
Program, Memorial Sloan Kettering Cancer
Center, 1275 York Avenue, New York, New York 10065, United States
| | - Yi Tang
- Department
of Chemistry and Biochemistry, UCLA, 607 Charles E. Young Drive East, Los Angeles, California 90095, United States
| | - Richard
E. Taylor
- Department
of Chemistry and Biochemistry and the Warren Family Research Center
for Drug Discovery and Development, University
of Notre Dame, 305 McCourtney
Hall, Notre Dame, Indiana 46556, United States
| | - Regan
J. Thomson
- Department
of Chemistry, Northwestern University, 2145 Sheridan Road, Evanston, Illinois 60208, United States
| | - David
A. Vosburg
- Department
of Chemistry, Harvey Mudd College, 301 Platt Boulevard, Claremont, California 91711, United States
| | - Jimmy Wu
- Department
of Chemistry, Dartmouth College, Hanover, New Hampshire 03755, United States
| | - William
M. Wuest
- Department
of Chemistry, Emory University, 1515 Dickey Drive, Atlanta, Georgia 30322, United States
- Emory Antibiotic
Resistance Center, Emory University School
of Medicine, 201 Dowman
Drive, Atlanta, Georgia 30322, United States
| | - Armen Zakarian
- Santa
Barbara
Department of Chemistry and Biochemistry, University of California, Santa
Barbara, California 93106, United States
| | - Yufeng Zhang
- School of
Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Sha Tin, New Territories, Hong Kong SAR
| | - Tianjing Ren
- School of
Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Sha Tin, New Territories, Hong Kong SAR
| | - Zhong Zuo
- School of
Pharmacy, Faculty of Medicine, The Chinese
University of Hong Kong, Sha Tin, New Territories, Hong Kong SAR
| | - James Inglese
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Sam Michael
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Anton Simeonov
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Wei Zheng
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Paul Shinn
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Ajit Jadhav
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Matthew
B. Boxer
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Matthew D. Hall
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Menghang Xia
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Rajarshi Guha
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| | - Jason M. Rohde
- National
Center for Advancing Translational Sciences, National Institutes of
Health, 9800 Medical Center Drive, Rockville, Maryland 20850, United States
| |
Collapse
|
32
|
Burke LC, Ezeribe HO, Kwon AY, Dockery D, Lyons PJ. Carboxypeptidase O is a lipid droplet-associated enzyme able to cleave both acidic and polar C-terminal amino acids. PLoS One 2018; 13:e0206824. [PMID: 30388170 PMCID: PMC6214572 DOI: 10.1371/journal.pone.0206824] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2017] [Accepted: 10/19/2018] [Indexed: 11/18/2022] Open
Abstract
Carboxypeptidase O (CPO) is a member of the M14 family of metallocarboxypeptidases with a preference for the cleavage of C-terminal acidic amino acids. CPO is largely expressed in the small intestine, although it has been detected in other tissues such as the brain and ovaries. CPO does not contain a prodomain, nor is it strongly regulated by pH, and hence appears to exist as a constitutively active enzyme. The goal of this study was to investigate the intracellular distribution and activity of CPO in order to predict physiological substrates and function. The distribution of CPO, when expressed in MDCK cells, was analyzed by immunofluorescence microscopy. Soon after addition of nutrient-rich media, CPO was found to associate with lipid droplets, causing an increase in lipid droplet quantity. As media became depleted, CPO moved to a broader ER distribution, no longer impacting lipid droplet numbers. Membrane cholesterol levels played a role in the distribution and in vitro enzymatic activity of CPO, with cholesterol enrichment leading to decreased lipid droplet association and enzymatic activity. The ability of CPO to cleave C-terminal amino acids within the early secretory pathway (in vivo) was examined using Gaussia luciferase as a substrate, C-terminally tagged with variants of an ER retention signal. While no effect of cholesterol was observed, these data show that CPO does function as an active enzyme within the ER where it removes C-terminal glutamates and aspartates, as well as a number of polar amino acids.
Collapse
Affiliation(s)
- Linnea C. Burke
- Department of Biology, Andrews University, Berrien Springs, Michigan, United States of America
| | - Hazel O. Ezeribe
- Department of Biology, Andrews University, Berrien Springs, Michigan, United States of America
| | - Anna Y. Kwon
- Department of Biology, Andrews University, Berrien Springs, Michigan, United States of America
| | - Donnel Dockery
- Department of Biology, Andrews University, Berrien Springs, Michigan, United States of America
| | - Peter J. Lyons
- Department of Biology, Andrews University, Berrien Springs, Michigan, United States of America
- * E-mail:
| |
Collapse
|
33
|
Abstract
Ryanodine receptor type 1-related myopathies (RYR1-RM) are the most common class of congenital myopathies. Historically, RYR1-RM classification and diagnosis have been guided by histopathologic findings on muscle biopsy. Main histological subtypes of RYR1-RM include central core disease, multiminicore disease, core-rod myopathy, centronuclear myopathy, and congenital fiber-type disproportion. A range of RYR1-RM clinical phenotypes has also emerged more recently and includes King Denborough syndrome, RYR1 rhabdomyolysis-myalgia syndrome, atypical periodic paralysis, congenital neuromuscular disease with uniform type 1 fibers, and late-onset axial myopathy. This expansion of the RYR1-RM disease spectrum is due, in part, to implementation of next-generation sequencing methods, which include the entire RYR1 coding sequence rather than being restricted to hotspot regions. These methods enhance diagnostic capabilities, especially given historic limitations of histopathologic and clinical overlap across RYR1-RM. Both dominant and recessive modes of inheritance have been documented, with the latter typically associated with a more severe clinical phenotype. As with all congenital myopathies, no FDA-approved treatments exist to date. Here, we review histopathologic, clinical, imaging, and genetic diagnostic features of the main RYR1-RM subtypes. We also discuss the current state of treatments and focus on disease-modulating (nongenetic) therapeutic strategies under development for RYR1-RM. Finally, perspectives for future approaches to treatment development are broached.
Collapse
Affiliation(s)
- Tokunbor A Lawal
- Neuromuscular Symptoms Unit, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA
| | - Joshua J Todd
- Neuromuscular Symptoms Unit, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA
| | - Katherine G Meilleur
- Neuromuscular Symptoms Unit, National Institute of Nursing Research, National Institutes of Health, Bethesda, MD, USA.
| |
Collapse
|
34
|
Trychta KA, Heathward EJ, Sulima A, Bäck S, Farokhnia M, Richie CT, Leggio L, Rice KC, Harvey BK. Extracellular esterase activity as an indicator of endoplasmic reticulum calcium depletion. Biomarkers 2018; 23:756-765. [PMID: 30095301 DOI: 10.1080/1354750x.2018.1490968] [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] [Indexed: 01/27/2023]
Abstract
CONTEXT Endoplasmic reticulum (ER) calcium depletion is associated with diverse diseases, including cardiac, hepatic, and neurologic diseases. OBJECTIVE The aim of the present study was to identify and characterize an endogenous protein that could be used to monitor ER calcium depletion comparably to a previously described exogenous reporter protein. MATERIALS AND METHODS The use of a selective esterase-fluorescein diester pair allowed for carboxylesterase activity in extracellular fluid to be measured using a fluorescent readout. Cell culture media from three different cell lines, rat plasma, and human serum all possess quantifiable amounts of esterase activity. RESULTS Fluorescence produced by the interaction of carboxylesterases with a fluorescein diester substrate tracks with pharmacological and physiological inducers of ER calcium depletion. The fluorescence measured for in vitro and in vivo samples were consistent with ER calcium depletion being the trigger for increased esterase activity. DISCUSSION Decreased luminal ER calcium causes ER resident esterases to be released from the cell, and, when assessed concurrently with other disease biomarkers, these esterases may provide insight into the role of ER calcium homeostasis in human diseases. CONCLUSION Our results indicate that carboxylesterases are putative markers of ER calcium dysfunction.
Collapse
Affiliation(s)
- Kathleen A Trychta
- a Molecular Mechanisms of Cellular Stress and Inflammation , Intramural Research Program, National Institute on Drug Abuse , Baltimore , MD , USA
| | - Emily J Heathward
- a Molecular Mechanisms of Cellular Stress and Inflammation , Intramural Research Program, National Institute on Drug Abuse , Baltimore , MD , USA
| | - Agnieszka Sulima
- b Section on Drug Design and Synthesis, Molecular Targets and Medications Branch , National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health , Bethesda , MD , USA
| | - Susanne Bäck
- a Molecular Mechanisms of Cellular Stress and Inflammation , Intramural Research Program, National Institute on Drug Abuse , Baltimore , MD , USA
| | - Mehdi Farokhnia
- c Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology , National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health , Bethesda , MD , USA
| | - Christopher T Richie
- a Molecular Mechanisms of Cellular Stress and Inflammation , Intramural Research Program, National Institute on Drug Abuse , Baltimore , MD , USA
| | - Lorenzo Leggio
- c Section on Clinical Psychoneuroendocrinology and Neuropsychopharmacology , National Institute on Alcohol Abuse and Alcoholism and National Institute on Drug Abuse, National Institutes of Health , Bethesda , MD , USA.,d Center for Alcohol and Addiction Studies, Department of Behavioral and Social Sciences , Brown University , Providence , RI , USA
| | - Kenner C Rice
- b Section on Drug Design and Synthesis, Molecular Targets and Medications Branch , National Institute on Drug Abuse and National Institute on Alcohol Abuse and Alcoholism, National Institutes of Health , Bethesda , MD , USA
| | - Brandon K Harvey
- a Molecular Mechanisms of Cellular Stress and Inflammation , Intramural Research Program, National Institute on Drug Abuse , Baltimore , MD , USA
| |
Collapse
|
35
|
Penttinen AM, Parkkinen I, Voutilainen MH, Koskela M, Bäck S, Their A, Richie CT, Domanskyi A, Harvey BK, Tuominen RK, Nevalaita L, Saarma M, Airavaara M. Pre-α-pro-GDNF and Pre-β-pro-GDNF Isoforms Are Neuroprotective in the 6-hydroxydopamine Rat Model of Parkinson's Disease. Front Neurol 2018; 9:457. [PMID: 29973907 PMCID: PMC6019446 DOI: 10.3389/fneur.2018.00457] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 05/29/2018] [Indexed: 11/13/2022] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) is one of the most studied neurotrophic factors. GDNF has two splice isoforms, full-length pre-α-pro-GDNF (α-GDNF) and pre-β-pro-GDNF (β-GDNF), which has a 26 amino acid deletion in the pro-region. Thus far, studies have focused solely on the α-GDNF isoform, and nothing is known about the in vivo effects of the shorter β-GDNF variant. Here we compare for the first time the effects of overexpressed α-GDNF and β-GDNF in non-lesioned rat striatum and the partial 6-hydroxydopamine lesion model of Parkinson's disease. GDNF isoforms were overexpressed with their native pre-pro-sequences in the striatum using an adeno-associated virus (AAV) vector, and the effects on motor performance and dopaminergic phenotype of the nigrostriatal pathway were assessed. In the non-lesioned striatum, both isoforms increased the density of dopamine transporter-positive fibers at 3 weeks after viral vector delivery. Although both isoforms increased the activity of the animals in cylinder assay, only α-GDNF enhanced the use of contralateral paw. Four weeks later, the striatal tyrosine hydroxylase (TH)-immunoreactivity was decreased in both α-GDNF and β-GDNF treated animals. In the neuroprotection assay, both GDNF splice isoforms increased the number of TH-immunoreactive cells in the substantia nigra but did not promote behavioral recovery based on amphetamine-induced rotation or cylinder assays. Thus, the shorter GDNF isoform, β-GDNF, and the full-length α-isoform have comparable neuroprotective efficacy on dopamine neurons of the nigrostriatal circuitry.
Collapse
Affiliation(s)
- Anna-Maija Penttinen
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Ilmari Parkkinen
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Merja H Voutilainen
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Maryna Koskela
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Susanne Bäck
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Anna Their
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Christopher T Richie
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Andrii Domanskyi
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Brandon K Harvey
- National Institute on Drug Abuse, National Institutes of Health, Baltimore, MD, United States
| | - Raimo K Tuominen
- Division of Pharmacology and Pharmacotherapy, Faculty of Pharmacy, University of Helsinki, Helsinki, Finland
| | - Liina Nevalaita
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mart Saarma
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| | - Mikko Airavaara
- HiLIFE Unit, Institute of Biotechnology, University of Helsinki, Helsinki, Finland
| |
Collapse
|
36
|
Campbell LA, Richie CT, Zhang Y, Heathward EJ, Coke LM, Park EY, Harvey BK. In vitro modeling of HIV proviral activity in microglia. FEBS J 2017; 284:4096-4114. [PMID: 29114997 DOI: 10.1111/febs.14293] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2017] [Revised: 09/14/2017] [Accepted: 10/13/2017] [Indexed: 12/19/2022]
Abstract
Microglia, the resident macrophages of the brain, play a key role in the pathogenesis of HIV-associated neurocognitive disorders (HAND) due to their productive infection by HIV. This results in the release of neurotoxic viral proteins and pro-inflammatory compounds which negatively affect the functionality of surrounding neurons. Because models of HIV infection within the brain are limited, we aimed to create a novel microglia cell line with an integrated HIV provirus capable of recreating several hallmarks of HIV infection. We utilized clustered regularly interspaced short palindromic repeats (CRISPR)/Cas9 gene editing technology and integrated a modified HIV provirus into CHME-5 immortalized microglia to create HIV-NanoLuc CHME-5. In the modified provirus, the Gag-Pol region is replaced with the coding region for NanoLuciferase (NanoLuc), which allows for the rapid assay of HIV long terminal repeat activity using a luminescent substrate, while still containing the necessary genetic material to produce established neurotoxic viral proteins (e.g. tat, nef, gp120). We confirmed that HIV-NanoLuc CHME-5 microglia express NanoLuc, along with the HIV viral protein Nef. We subsequently exposed these cells to a battery of experiments to modulate the activity of the provirus. Proviral activity was enhanced by treating the cells with pro-inflammatory factors lipopolysaccharide (LPS) and tumor necrosis factor alpha and by overexpressing the viral regulatory protein Tat. Conversely, genetic modification of the toll-like receptor-4 gene by CRISPR/Cas9 reduced LPS-mediated proviral activation, and pharmacological application of NF-κB inhibitor sulfasalazine similarly diminished proviral activity. Overall, these data suggest that HIV-NanoLuc CHME-5 may be a useful tool in the study of HIV-mediated neuropathology and proviral regulation.
Collapse
Affiliation(s)
- Lee A Campbell
- Intramural Research Program, National Institute on Drug Abuse, Biomedical Research Center, Baltimore, MD, USA
| | - Christopher T Richie
- Intramural Research Program, National Institute on Drug Abuse, Biomedical Research Center, Baltimore, MD, USA
| | - Yajun Zhang
- Intramural Research Program, National Institute on Drug Abuse, Biomedical Research Center, Baltimore, MD, USA
| | - Emily J Heathward
- Intramural Research Program, National Institute on Drug Abuse, Biomedical Research Center, Baltimore, MD, USA
| | - Lamarque M Coke
- Intramural Research Program, National Institute on Drug Abuse, Biomedical Research Center, Baltimore, MD, USA
| | - Emily Y Park
- Intramural Research Program, National Institute on Drug Abuse, Biomedical Research Center, Baltimore, MD, USA
| | - Brandon K Harvey
- Intramural Research Program, National Institute on Drug Abuse, Biomedical Research Center, Baltimore, MD, USA
| |
Collapse
|
37
|
High fat diet disrupts endoplasmic reticulum calcium homeostasis in the rat liver. J Hepatol 2017; 67:1009-1017. [PMID: 28596111 PMCID: PMC6122848 DOI: 10.1016/j.jhep.2017.05.023] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 05/10/2017] [Accepted: 05/25/2017] [Indexed: 12/14/2022]
Abstract
BACKGROUND & AIMS Disruption to endoplasmic reticulum (ER) calcium homeostasis has been implicated in obesity, however, the ability to longitudinally monitor ER calcium fluctuations has been challenging with prior methodologies. We recently described the development of a Gaussia luciferase (GLuc)-based reporter protein responsive to ER calcium depletion (GLuc-SERCaMP) and investigated the effect of a high fat diet on ER calcium homeostasis. METHODS A GLuc-based reporter cell line was treated with palmitate, a free fatty acid. Rats intrahepatically injected with GLuc-SERCaMP reporter were fed a cafeteria diet or high fat diet. The liver and plasma were examined for established markers of steatosis and compared to plasma levels of SERCaMP activity. RESULTS Palmitate induced GLuc-SERCaMP release in vitro, indicating ER calcium depletion. Consumption of a cafeteria diet or high fat pellets correlated with alterations to hepatic ER calcium homeostasis in rats, shown by increased GLuc-SERCaMP release. Access to ad lib high fat pellets also led to a corresponding decrease in microsomal calcium ATPase activity and an increase in markers of hepatic steatosis. In addition to GLuc-SERCaMP, we have also identified endogenous proteins (endogenous SERCaMPs) with a similar response to ER calcium depletion. We demonstrated the release of an endogenous SERCaMP, thought to be a liver esterase, during access to a high fat diet. Attenuation of both GLuc-SERCaMP and endogenous SERCaMP was observed during dantrolene administration. CONCLUSIONS Here we describe the use of a reporter for in vitro and in vivo models of high fat diet. Our results support the theory that dietary fat intake correlates with a decrease in ER calcium levels in the liver and suggest a high fat diet alters the ER proteome. Lay summary: ER calcium dysregulation was observed in rats fed a cafeteria diet or high fat pellets, with fluctuations in sensor release correlating with fat intake. Attenuation of sensor release, as well as food intake was observed during administration of dantrolene, a drug that stabilizes ER calcium. The study describes a novel technique for liver research and provides insight into cellular processes that may contribute to the pathogenesis of obesity and fatty liver disease.
Collapse
|
38
|
Sharpe MJ, Marchant NJ, Whitaker LR, Richie CT, Zhang YJ, Campbell EJ, Koivula PP, Necarsulmer JC, Mejias-Aponte C, Morales M, Pickel J, Smith JC, Niv Y, Shaham Y, Harvey BK, Schoenbaum G. Lateral Hypothalamic GABAergic Neurons Encode Reward Predictions that Are Relayed to the Ventral Tegmental Area to Regulate Learning. Curr Biol 2017; 27:2089-2100.e5. [PMID: 28690111 PMCID: PMC5564224 DOI: 10.1016/j.cub.2017.06.024] [Citation(s) in RCA: 88] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2017] [Revised: 05/08/2017] [Accepted: 06/09/2017] [Indexed: 12/27/2022]
Abstract
Eating is a learned process. Our desires for specific foods arise through experience. Both electrical stimulation and optogenetic studies have shown that increased activity in the lateral hypothalamus (LH) promotes feeding. Current dogma is that these effects reflect a role for LH neurons in the control of the core motivation to feed, and their activity comes under control of forebrain regions to elicit learned food-motivated behaviors. However, these effects could also reflect the storage of associative information about the cues leading to food in LH itself. Here, we present data from several studies that are consistent with a role for LH in learning. In the first experiment, we use a novel GAD-Cre rat to show that optogenetic inhibition of LH γ-aminobutyric acid (GABA) neurons restricted to cue presentation disrupts the rats' ability to learn that a cue predicts food without affecting subsequent food consumption. In the second experiment, we show that this manipulation also disrupts the ability of a cue to promote food seeking after learning. Finally, we show that inhibition of the terminals of the LH GABA neurons in ventral-tegmental area (VTA) facilitates learning about reward-paired cues. These results suggest that the LH GABA neurons are critical for storing and later disseminating information about reward-predictive cues.
Collapse
Affiliation(s)
- Melissa J Sharpe
- National Institute on Drug Abuse, IRP, 251 Bayview Boulevard, Baltimore, MD 21228, USA; Princeton Neuroscience Institute, Princeton University, Washington Road, Princeton, NJ 08544, USA.
| | - Nathan J Marchant
- National Institute on Drug Abuse, IRP, 251 Bayview Boulevard, Baltimore, MD 21228, USA; Florey Institute of Neuroscience and Mental Health, University of Melbourne, 30 Royal Parade, Melbourne, VIC 3052, Australia
| | - Leslie R Whitaker
- National Institute on Drug Abuse, IRP, 251 Bayview Boulevard, Baltimore, MD 21228, USA
| | - Christopher T Richie
- National Institute on Drug Abuse, IRP, 251 Bayview Boulevard, Baltimore, MD 21228, USA
| | - Yajun J Zhang
- National Institute on Drug Abuse, IRP, 251 Bayview Boulevard, Baltimore, MD 21228, USA; National Institute on Alcohol Abuse and Alcoholism, IRP, Executive Boulevard No. 402, Rockville, MD 20852, USA
| | - Erin J Campbell
- Neurobiology of Addiction Laboratory, School of Biomedical Sciences and Pharmacy, University of Newcastle and the Hunter Medical Research Institute, University Drive, Newcastle, NSW 2308, Australia
| | - Pyry P Koivula
- National Institute on Drug Abuse, IRP, 251 Bayview Boulevard, Baltimore, MD 21228, USA
| | - Julie C Necarsulmer
- National Institute on Drug Abuse, IRP, 251 Bayview Boulevard, Baltimore, MD 21228, USA
| | - Carlos Mejias-Aponte
- National Institute on Drug Abuse, IRP, 251 Bayview Boulevard, Baltimore, MD 21228, USA
| | - Marisela Morales
- National Institute on Drug Abuse, IRP, 251 Bayview Boulevard, Baltimore, MD 21228, USA
| | - James Pickel
- National Institute of Mental Health, IRP, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Jeffrey C Smith
- National Institute of Neurological Disorders and Stroke, IRP, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | - Yael Niv
- Princeton Neuroscience Institute, Princeton University, Washington Road, Princeton, NJ 08544, USA
| | - Yavin Shaham
- National Institute on Drug Abuse, IRP, 251 Bayview Boulevard, Baltimore, MD 21228, USA
| | - Brandon K Harvey
- National Institute on Drug Abuse, IRP, 251 Bayview Boulevard, Baltimore, MD 21228, USA.
| | - Geoffrey Schoenbaum
- National Institute on Drug Abuse, IRP, 251 Bayview Boulevard, Baltimore, MD 21228, USA; Department of Anatomy and Neurobiology, University of Maryland School of Medicine, 20 Penn Street, Baltimore, MD 21201, USA; Solomon H. Snyder Department of Neuroscience, John Hopkins University, 401 N. Broadway, Baltimore, MD 21287, USA.
| |
Collapse
|
39
|
Mesencephalic astrocyte-derived neurotrophic factor and its role in nervous system disease. Neurol Sci 2017; 38:1741-1746. [DOI: 10.1007/s10072-017-3042-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2016] [Accepted: 06/19/2017] [Indexed: 12/22/2022]
|
40
|
Howard DB, Harvey BK. Assaying the Stability and Inactivation of AAV Serotype 1 Vectors. Hum Gene Ther Methods 2017; 28:39-48. [PMID: 28192678 DOI: 10.1089/hgtb.2016.180] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Adeno-associated virus (AAV) vectors are a commonplace tool for gene delivery ranging from cell culture to human gene therapy. One feature that makes AAV a desirable vector is its stability, in regard to both the duration of transgene expression and retention of infectivity as a viral particle. This study examined the stability of AAV serotype 1 (AAV1) vectors under different conditions. First, transducibility after storage at 4°C decreased 20% over 7 weeks. Over 10 freeze-thaw cycles, the resulting transduction efficiency became variable at 60-120% of a single thaw. Using small stainless steel slugs to mimic a biosafety cabinet or metal lab bench surface, it was found that an AAV1 vector can be reconstituted after 6 days of storage at room temperature. The stability of AAV is a desired feature, but effective decontamination procedures must be available for safety and experimental integrity. Multiple disinfectants commonly used in the laboratory for ability to inactivate an AAV1 vector were tested, and it was found that autoclaving, 0.25% peracetic acid, iodine, or 10% Clorox bleach completely prevented AAV-mediated transgene expression. These data suggest that peracetic acid should be used for inactivating AAV1 vectors on metal-based surfaces or instruments in order to avoid inadvertent transgene expression in human cells or cross-contamination of instruments.
Collapse
Affiliation(s)
- Douglas B Howard
- Intramural Research Program, National Institute on Drug Abuse , Baltimore, Maryland
| | - Brandon K Harvey
- Intramural Research Program, National Institute on Drug Abuse , Baltimore, Maryland
| |
Collapse
|
41
|
Richie CT, Whitaker LR, Whitaker KW, Necarsulmer J, Baldwin HA, Zhang Y, Fortuno L, Hinkle JJ, Koivula P, Henderson MJ, Sun W, Wang K, Smith JC, Pickel J, Ji N, Hope BT, Harvey BK. Near-infrared fluorescent protein iRFP713 as a reporter protein for optogenetic vectors, a transgenic Cre-reporter rat, and other neuronal studies. J Neurosci Methods 2017; 284:1-14. [PMID: 28380331 PMCID: PMC5501963 DOI: 10.1016/j.jneumeth.2017.03.020] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2017] [Revised: 03/29/2017] [Accepted: 03/31/2017] [Indexed: 01/21/2023]
Abstract
BACKGROUND The use of genetically-encoded fluorescent reporters is essential for the identification and observation of cells that express transgenic modulatory proteins. Near-infrared (NIR) fluorescent proteins have superior light penetration through biological tissue, but are not yet widely adopted. NEW METHOD Using the near-infrared fluorescent protein, iRFP713, improves the imaging resolution in thick tissue sections or the intact brain due to the reduced light-scattering at the longer, NIR wavelengths used to image the protein. Additionally, iRFP713 can be used to identify transgenic cells without photobleaching other fluorescent reporters or affecting opsin function. We have generated a set of adeno-associated vectors in which iRFP713 has been fused to optogenetic channels, and can be expressed constitutively or Cre-dependently. RESULTS iRFP713 is detectable when expressed in neurons both in vitro and in vivo without exogenously supplied chromophore biliverdin. Neuronally-expressed iRFP713 has similar properties to GFP-like fluorescent proteins, including the ability to be translationally fused to channelrhodopsin or halorhodopsin, however, it shows superior photostability compared to EYFP. Furthermore, electrophysiological recordings from iRFP713-labeled cells compared to cells labeled with mCherry suggest that iRFP713 cells are healthier and therefore more stable and reliable in an ex vivo preparation. Lastly, we have generated a transgenic rat that expresses iRFP713 in a Cre-dependent manner. CONCLUSIONS Overall, we have demonstrated that iRFP713 can be used as a reporter in neurons without the use of exogenous biliverdin, with minimal impact on viability and function thereby making it feasible to extend the capabilities for imaging genetically-tagged neurons in slices and in vivo.
Collapse
Affiliation(s)
- Christopher T Richie
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, United States
| | - Leslie R Whitaker
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, United States
| | - Keith W Whitaker
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, United States; US Army Research Laboratory, Aberdeen Proving Ground, MD 21005, United States
| | - Julie Necarsulmer
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, United States
| | - Heather A Baldwin
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, United States
| | - Yajun Zhang
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, United States; Intramural Research Program, National Institute on Alcohol Abuse and Alcoholism, Rockville, MD 20852, United States
| | - Lowella Fortuno
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, United States
| | - Josh J Hinkle
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, United States
| | - Pyry Koivula
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, United States
| | - Mark J Henderson
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, United States
| | - Wenzhi Sun
- Janelia Research Campus,Howard Hughes Medical Institute, Ashburn, VA 20147, United States
| | - Kai Wang
- Janelia Research Campus,Howard Hughes Medical Institute, Ashburn, VA 20147, United States
| | - Jeffrey C Smith
- Intramural Research Program, National Institute of Neurological Disorders and Stroke, Bethesda, MD 20892, United States
| | - Jim Pickel
- Intramural Research Program, National Institute of Mental Health, Bethesda, MD 20892, United States
| | - Na Ji
- Janelia Research Campus,Howard Hughes Medical Institute, Ashburn, VA 20147, United States
| | - Bruce T Hope
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, United States
| | - Brandon K Harvey
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, MD 21224, United States.
| |
Collapse
|
42
|
Wires ES, Henderson MJ, Yan X, Bäck S, Trychta KA, Lutrey MH, Harvey BK. Longitudinal monitoring of Gaussia and Nano luciferase activities to concurrently assess ER calcium homeostasis and ER stress in vivo. PLoS One 2017; 12:e0175481. [PMID: 28403212 PMCID: PMC5389830 DOI: 10.1371/journal.pone.0175481] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2016] [Accepted: 03/27/2017] [Indexed: 12/13/2022] Open
Abstract
The endoplasmic reticulum (ER) is essential to many cellular processes including protein processing, lipid metabolism and calcium storage. The ability to longitudinally monitor ER homeostasis in the same organism would offer insight into progressive molecular and cellular adaptations to physiologic or pathologic states, but has been challenging. We recently described the creation of a Gaussia luciferase (GLuc)-based secreted ER calcium-modulated protein (SERCaMP or GLuc-SERCaMP) to longitudinally monitor ER calcium homeostasis. Here we describe a complementary tool to measure the unfolded protein response (UPR), utilizing a UPRE-driven secreted Nano luciferase (UPRE-secNLuc) to examine the activating transcription factor-6 (ATF6) and inositol-requiring enzyme 1 (IRE1) pathways of the UPR. We observed an upregulation of endogenous ATF6- and XBP1-regulated genes following pharmacologically-induced ER stress that was consistent with responsiveness of the UPRE sensor. Both GLuc and NLuc-based reporters have favorable properties for in vivo studies, however, they are not easily used in combination due to overlapping substrate activities. We describe a method to measure the enzymatic activities of both reporters from a single sample and validated the approach using culture medium and rat blood samples to measure GLuc-SERCaMP and UPRE-secNLuc. Measuring GLuc and NLuc activities from the same sample allows for the robust and quantitative measurement of two cellular events or cell populations from a single biological sample. This study is the first to describe the in vivo measurement of UPRE activation by sampling blood, using an approach that allows concurrent interrogation of two components of ER homeostasis.
Collapse
Affiliation(s)
- Emily S. Wires
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, United States of America
| | - Mark J. Henderson
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, United States of America
| | - Xiaokang Yan
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, United States of America
| | - Susanne Bäck
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, United States of America
| | - Kathleen A. Trychta
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, United States of America
| | - Molly H. Lutrey
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, United States of America
| | - Brandon K. Harvey
- Intramural Research Program, National Institute on Drug Abuse, Baltimore, Maryland, United States of America
- * E-mail:
| |
Collapse
|
43
|
Lindahl M, Saarma M, Lindholm P. Unconventional neurotrophic factors CDNF and MANF: Structure, physiological functions and therapeutic potential. Neurobiol Dis 2016; 97:90-102. [PMID: 27425895 DOI: 10.1016/j.nbd.2016.07.009] [Citation(s) in RCA: 125] [Impact Index Per Article: 13.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2016] [Revised: 06/29/2016] [Accepted: 07/13/2016] [Indexed: 12/30/2022] Open
Abstract
Cerebral dopamine neurotrophic factor (CDNF) and mesencephalic astrocyte-derived neurotrophic factor (MANF) promote the survival of midbrain dopaminergic neurons which degenerate in Parkinson's disease (PD). However, CDNF and MANF are structurally and functionally clearly distinct from the classical, target-derived neurotrophic factors (NTFs) that are solely secreted proteins. In cells, CDNF and MANF localize in the endoplasmic reticulum (ER) and evidence suggests that MANF, and possibly CDNF, is important for the maintenance of ER homeostasis. MANF expression is particularly high in secretory tissues with extensive protein production and thus a high ER protein folding load. Deletion of MANF in mice results in a diabetic phenotype and the activation of unfolded protein response (UPR) in the pancreatic islets. However, information about the intracellular and extracellular mechanisms of MANF and CDNF action is still limited. Here we will discuss the structural motifs and physiological functions of CDNF and MANF as well as their therapeutic potential for the treatment of neurodegenerative diseases and diabetes. Currently available knockout models of MANF and CDNF in mice, zebrafish and fruit fly will increase information about the biology of these interesting proteins.
Collapse
Affiliation(s)
- Maria Lindahl
- Institute of Biotechnology, P.O.Box 56, Viikinkaari 5, FI-00014, University of Helsinki, Finland
| | - Mart Saarma
- Institute of Biotechnology, P.O.Box 56, Viikinkaari 5, FI-00014, University of Helsinki, Finland
| | - Päivi Lindholm
- Institute of Biotechnology, P.O.Box 56, Viikinkaari 5, FI-00014, University of Helsinki, Finland.
| |
Collapse
|
44
|
Role of two sequence motifs of mesencephalic astrocyte-derived neurotrophic factor in its survival-promoting activity. Cell Death Dis 2015; 6:e2032. [PMID: 26720341 PMCID: PMC4720903 DOI: 10.1038/cddis.2015.371] [Citation(s) in RCA: 45] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2014] [Revised: 11/17/2015] [Accepted: 11/19/2015] [Indexed: 12/21/2022]
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) is a prosurvival protein that protects the cells when applied intracellularly in vitro or extracellularly in vivo. Its protective mechanisms are poorly known. Here we studied the role of two short sequence motifs within the carboxy-(C) terminal domain of MANF in its neuroprotective activity: the CKGC sequence (a CXXC motif) that could be involved in redox reactions, and the C-terminal RTDL sequence, an endoplasmic reticulum (ER) retention signal. We mutated these motifs and analyzed the antiapoptotic effect and intracellular localization of these mutants of MANF when overexpressed in cultured sympathetic or sensory neurons. As an in vivo model for studying the effect of these mutants after their extracellular application, we used the rat model of cerebral ischemia. Even though we found no evidence for oxidoreductase activity of MANF, the mutation of CXXC motif completely abolished its protective effect, showing that this motif is crucial for both MANF's intracellular and extracellular activity. The RTDL motif was not needed for the neuroprotective activity of MANF after its extracellular application in the stroke model in vivo. However, in vitro the deletion of RTDL motif inactivated MANF in the sympathetic neurons where the mutant protein localized to Golgi, but not in the sensory neurons where the mutant localized to the ER, showing that intracellular MANF protects these peripheral neurons in vitro only when localized to the ER.
Collapse
|
45
|
Henderson MJ, Baldwin HA, Werley CA, Boccardo S, Whitaker LR, Yan X, Holt GT, Schreiter ER, Looger LL, Cohen AE, Kim DS, Harvey BK. A Low Affinity GCaMP3 Variant (GCaMPer) for Imaging the Endoplasmic Reticulum Calcium Store. PLoS One 2015; 10:e0139273. [PMID: 26451944 PMCID: PMC4599735 DOI: 10.1371/journal.pone.0139273] [Citation(s) in RCA: 49] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/12/2015] [Accepted: 09/09/2015] [Indexed: 12/22/2022] Open
Abstract
Endoplasmic reticulum calcium homeostasis is critical for cellular functions and is disrupted in diverse pathologies including neurodegeneration and cardiovascular disease. Owing to the high concentration of calcium within the ER, studying this subcellular compartment requires tools that are optimized for these conditions. To develop a single-fluorophore genetically encoded calcium indicator for this organelle, we targeted a low affinity variant of GCaMP3 to the ER lumen (GCaMPer (10.19)). A set of viral vectors was constructed to express GCaMPer in human neuroblastoma cells, rat primary cortical neurons, and human induced pluripotent stem cell-derived cardiomyocytes. We observed dynamic changes in GCaMPer (10.19) fluorescence in response to pharmacologic manipulations of the ER calcium store. Additionally, periodic calcium efflux from the ER was observed during spontaneous beating of cardiomyocytes. GCaMPer (10.19) has utility in imaging ER calcium in living cells and providing insight into luminal calcium dynamics under physiologic and pathologic states.
Collapse
Affiliation(s)
- Mark J. Henderson
- National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd, Baltimore, Maryland, 21224, United States of America
- * E-mail: (MJH); (BKH)
| | - Heather A. Baldwin
- National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd, Baltimore, Maryland, 21224, United States of America
| | - Christopher A. Werley
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, 02138, United States of America
| | - Stefano Boccardo
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, 02138, United States of America
| | - Leslie R. Whitaker
- National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd, Baltimore, Maryland, 21224, United States of America
| | - Xiaokang Yan
- National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd, Baltimore, Maryland, 21224, United States of America
| | - Graham T. Holt
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, Virginia, 20147, United States of America
| | - Eric R. Schreiter
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, Virginia, 20147, United States of America
| | - Loren L. Looger
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, Virginia, 20147, United States of America
| | - Adam E. Cohen
- Department of Chemistry and Chemical Biology, Harvard University, Cambridge, Massachusetts, 02138, United States of America
- Department of Physics, Harvard University, Cambridge, Massachusetts, 02138, United States of America
- Harvard Stem Cell Institute, Harvard University, Cambridge, Massachusetts, 02138, United States of America
- Howard Hughes Medical Institute, Harvard University, Cambridge, Massachusetts, 02138, United States of America
| | - Douglas S. Kim
- Janelia Research Campus, Howard Hughes Medical Institute, 19700 Helix Drive, Ashburn, Virginia, 20147, United States of America
| | - Brandon K. Harvey
- National Institute on Drug Abuse, National Institutes of Health, 251 Bayview Blvd, Baltimore, Maryland, 21224, United States of America
- * E-mail: (MJH); (BKH)
| |
Collapse
|
46
|
Henderson MJ, Wires ES, Trychta KA, Yan X, Harvey BK. Monitoring Endoplasmic Reticulum Calcium Homeostasis Using a Gaussia Luciferase SERCaMP. J Vis Exp 2015. [PMID: 26383227 DOI: 10.3791/53199] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
The endoplasmic reticulum (ER) contains the highest level of intracellular calcium, with concentrations approximately 5,000-fold greater than cytoplasmic levels. Tight control over ER calcium is imperative for protein folding, modification and trafficking. Perturbations to ER calcium can result in the activation of the unfolded protein response, a three-prong ER stress response mechanism, and contribute to pathogenesis in a variety of diseases. The ability to monitor ER calcium alterations during disease onset and progression is important in principle, yet challenging in practice. Currently available methods for monitoring ER calcium, such as calcium-dependent fluorescent dyes and proteins, have provided insight into ER calcium dynamics in cells, however these tools are not well suited for in vivo studies. Our lab has demonstrated that a modification to the carboxy-terminus of Gaussia luciferase confers secretion of the reporter in response to ER calcium depletion. The methods for using a luciferase based, secreted ER calcium monitoring protein (SERCaMP) for in vitro and in vivo applications are described herein. This video highlights hepatic injections, pharmacological manipulation of GLuc-SERCaMP, blood collection and processing, and assay parameters for longitudinal monitoring of ER calcium.
Collapse
Affiliation(s)
| | - Emily S Wires
- National Institute on Drug Abuse, National Institutes of Health
| | | | - Xiaokang Yan
- National Institute on Drug Abuse, National Institutes of Health
| | - Brandon K Harvey
- National Institute on Drug Abuse, National Institutes of Health;
| |
Collapse
|
47
|
Liu H, Tang X, Gong L. Mesencephalic astrocyte-derived neurotrophic factor and cerebral dopamine neurotrophic factor: New endoplasmic reticulum stress response proteins. Eur J Pharmacol 2015; 750:118-22. [PMID: 25637781 DOI: 10.1016/j.ejphar.2015.01.016] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2014] [Revised: 12/09/2014] [Accepted: 01/07/2015] [Indexed: 01/21/2023]
Abstract
Mesencephalic astrocyte-derived neurotrophic factor (MANF) and cerebral dopamine neurotrophic factor (CDNF) are a novel evolutionary conserved neurotrophic factor (NTF) family. There are two distinct domains in MANF and CDNF 3-dimentional structure, N-terminal saposin-like domain and C-terminal SAP-domain, which suggest their unique mode of action. Although identified for their neurotrophic activity, recent studies have shown MANF and CDNF can protect cells during endoplasmic reticulum (ER) stress. This review summarizes the unique structure and related potential protective role for cells during ER stress of MANF and CDNF.
Collapse
Affiliation(s)
- Hao Liu
- Yuhuangding Hospital, Yantai, Shandong Province, PR China
| | - Xiaolei Tang
- Taishan Medical College, Taian, Shandong Province, PR China
| | - Lei Gong
- Yuhuangding Hospital, Yantai, Shandong Province, PR China.
| |
Collapse
|