1
|
Shokr MM, Badawi GA, Elshazly SM, Zaki HF, Mohamed AF. Sigma 1 Receptor and Its Pivotal Role in Neurological Disorders. ACS Pharmacol Transl Sci 2025; 8:47-65. [PMID: 39816800 PMCID: PMC11729429 DOI: 10.1021/acsptsci.4c00564] [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: 09/23/2024] [Revised: 12/07/2024] [Accepted: 12/16/2024] [Indexed: 01/18/2025]
Abstract
Sigma 1 receptor (S1R) is a multifunctional, ligand-activated protein located in the membranes of the endoplasmic reticulum (ER). It mediates a variety of neurological disorders, including epilepsy, amyotrophic lateral sclerosis, Alzheimer's disease, Huntington's disease. The wide neuroprotective effects of S1R agonists are achieved by a variety of pro-survival and antiapoptotic S1R-mediated signaling functions. Nonetheless, relatively little is known about the specific molecular mechanisms underlying S1R activity. Many studies on S1R protein have highlighted the importance of maintaining normal cellular homeostasis through its control of calcium and lipid exchange between the ER and mitochondria, ER-stress response, and many other mechanisms. In this review, we will discuss S1R different cellular localization and explain S1R-associated biological activity, such as its localization in the ER-plasma membrane and Mitochondrion-Associated ER Membrane interfaces. While outlining the cellular mechanisms and important binding partners involved in these processes, we also explained how the dysregulation of these pathways contributes to neurodegenerative disorders.
Collapse
Affiliation(s)
- Mustafa M. Shokr
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University−Arish Branch, Arish, 45511, Egypt
| | - Ghada A. Badawi
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Sinai University−Arish Branch, Arish, 45511, Egypt
| | - Shimaa M. Elshazly
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Zagazig University, Zagazig, Egypt
| | - Hala F. Zaki
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
| | - Ahmed F. Mohamed
- Department
of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Cairo, Egypt
- Faculty
of Pharmacy, King Salman International University
(KSIU), South Sinai 46612, Egypt
| |
Collapse
|
2
|
Song Y, Xu Z, Zhang L, Gao L. Sigma-1 Receptor Modulates CFA-Induced Inflammatory Pain via Sodium Channels in Small DRG Neurons. Biomolecules 2025; 15:73. [PMID: 39858467 PMCID: PMC11764217 DOI: 10.3390/biom15010073] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 01/03/2025] [Accepted: 01/05/2025] [Indexed: 01/30/2025] Open
Abstract
The sigma-1 receptor (Sig-1R) has emerged as a significant target in the realm of pain management and has been the subject of extensive research. Nonetheless, its specific function in inflammatory pain within dorsal root ganglion (DRG) neurons remains inadequately elucidated. This study utilized whole-cell patch clamp techniques, single-cell real-time PCR, and immunohistochemistry to examine the influence of Sig-1R on inflammatory pain induced by complete Freund's adjuvant (CFA) in a rat model. Our results revealed several key findings: (1) The expression of Sig-1R was found to be upregulated during the progression of inflammatory pain, with a notable translocation from the cytoplasm to the membrane; (2) Inhibition of peripheral Sig-1R using S1RA resulted in a reduction of CFA-induced allodynia; (3) Activation of Sig-1R through PRE-084 led to a decrease in the fast sodium current in isolated DRG neurons from CFA-treated rats, which was associated with a diminished action potential (AP) peak and maximum depolarizing rate (MDR), as well as an increased rheobase; (4) Furthermore, PRE-084 was observed to enhance the slow component of the sodium current, resulting in hyperpolarization of the threshold potential and an increase in AP firing frequency, alongside an elevation in the mRNA expression of the slow sodium channel Nav1.9 in CFA-treated rats. In conclusion, our findings suggest that the modulation of sodium channels by Sig-1R in DRG neurons plays a significant role in the mechanisms underlying inflammatory pain.
Collapse
Affiliation(s)
- Yuanlong Song
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China; (Y.S.); (Z.X.); (L.Z.)
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China
| | - Zifen Xu
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China; (Y.S.); (Z.X.); (L.Z.)
- College of Life Sciences and Health, Wuhan University of Science and Technology, Wuhan 430065, China
| | - Liangpin Zhang
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China; (Y.S.); (Z.X.); (L.Z.)
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China
| | - Linlin Gao
- Department of Physiology, School of Basic Medicine, Tongji Medical College, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China; (Y.S.); (Z.X.); (L.Z.)
- Hubei Key Laboratory of Drug Target Research and Pharmacodynamic Evaluation, Huazhong University of Science and Technology, 13 Hangkong Rd., Wuhan 430030, China
| |
Collapse
|
3
|
Soldatov V, Venediktov A, Belykh A, Piavchenko G, Naimzada MD, Ogneva N, Kartashkina N, Bushueva O. Chaperones vs. oxidative stress in the pathobiology of ischemic stroke. Front Mol Neurosci 2024; 17:1513084. [PMID: 39723236 PMCID: PMC11668803 DOI: 10.3389/fnmol.2024.1513084] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2024] [Accepted: 11/20/2024] [Indexed: 12/28/2024] Open
Abstract
As many proteins prioritize functionality over constancy of structure, a proteome is the shortest stave in the Liebig's barrel of cell sustainability. In this regard, both prokaryotes and eukaryotes possess abundant machinery supporting the quality of the proteome in healthy and stressful conditions. This machinery, namely chaperones, assists in folding, refolding, and the utilization of client proteins. The functions of chaperones are especially important for brain cells, which are highly sophisticated in terms of structural and functional organization. Molecular chaperones are known to exert beneficial effects in many brain diseases including one of the most threatening and widespread brain pathologies, ischemic stroke. However, whether and how they exert the antioxidant defense in stroke remains unclear. Herein, we discuss the chaperones shown to fight oxidative stress and the mechanisms of their antioxidant action. In ischemic stroke, during intense production of free radicals, molecular chaperones preserve the proteome by interacting with oxidized proteins, regulating imbalanced mitochondrial function, and directly fighting oxidative stress. For instance, cells recruit Hsp60 and Hsp70 to provide proper folding of newly synthesized proteins-these factors are required for early ischemic response and to refold damaged polypeptides. Additionally, Hsp70 upregulates some dedicated antioxidant pathways such as FOXO3 signaling. Small HSPs decrease oxidative stress via attenuation of mitochondrial function through their involvement in the regulation of Nrf- (Hsp22), Akt and Hippo (Hsp27) signaling pathways as well as mitophagy (Hsp27, Hsp22). A similar function has also been proposed for the Sigma-1 receptor, contributing to the regulation of mitochondrial function. Some chaperones can prevent excessive formation of reactive oxygen species whereas Hsp90 is suggested to be responsible for pro-oxidant effects in ischemic stroke. Finally, heat-resistant obscure proteins (Hero) are able to shield client proteins, thus preventing their possible over oxidation.
Collapse
Affiliation(s)
- Vladislav Soldatov
- Department of Pharmacology and Clinical Pharmacology, Belgorod State National Research University, Belgorod, Russia
| | - Artem Venediktov
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Andrei Belykh
- Pathophysiology Department, Kursk State Medical University, Kursk, Russia
- Research Institute of General Pathology, Kursk State Medical University, Kursk, Russia
| | - Gennadii Piavchenko
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Mukhammad David Naimzada
- Research Institute of Experimental Medicine, Kursk State Medical University, Kursk, Russia
- Laboratory of Public Health Indicators Analysis and Health Digitalization, Moscow Institute of Physics and Technology, Dolgoprudny, Russia
| | - Nastasya Ogneva
- Scientific Center of Biomedical Technologies, Federal Medical and Biological Agency of Russia, Moscow, Russia
| | - Natalia Kartashkina
- Department of Human Anatomy and Histology, I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow, Russia
| | - Olga Bushueva
- Laboratory of Genomic Research, Research Institute for Genetic and Molecular Epidemiology, Kursk State Medical University, Kursk, Russia
- Department of Biology, Medical Genetics and Ecology, Kursk State Medical University, Kursk, Russia
| |
Collapse
|
4
|
Yıldız B, Demirel R, Havadar HB, Yıldız G, Öziç C, Kamiloğlu NN, Özden Ö. Blocking SIG1R Along with Low Cadmium Exposure Display Anti-cancer Qualities in Both MCF7 and MDA-MB-231 Cells. Biol Trace Elem Res 2024; 202:3588-3600. [PMID: 37940833 DOI: 10.1007/s12011-023-03947-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/01/2023] [Accepted: 10/28/2023] [Indexed: 11/10/2023]
Abstract
Sigma-1 receptor (SIG1R) is a chaperone that modulates inositol 1,4,5-trisphosphate receptor type1 (IP3R1) calcium (Ca2+) channels on the endoplasmic reticulum. Therefore, SIG1R functions as an indirect regulator of Ca2+ and acts as an apoptosis modulator. Increased expression of SIG1R is associated with poor prognosis in breast cancers (BC), and SIG1R antagonists like BD1047 induce apoptosis. As a heavy metal, cadmium (Cd2+) is competitive with Ca2+ due to its physicochemical similarities and may trigger apoptosis at low concentrations. Our study investigated the SIG1R protein expression in 74 BC patients and found a significant increase in SIG1R expression in the triple-negative BC subtype. We also examined the apoptotic and anti-cancer effects of BD1047 in combination with CdCl2 in MCF7 and MDA-MB-213 cells. Cells were treated with CdCl2 at doses of 1 μM, 25 μM, and 50 μM, along with BD1047. Higher doses of CdCl2 were cytotoxic on both cancer cells and significantly increased DNA breaks. However, low-dose CdCl2 with BD1047 increased cell death and the apoptotic index in BC cells, although it did not exhibit cytotoxic effects on HUVEC cells. Co-administration of low-dose CdCl2 with BD1047 also reduced the migration and colony-forming ability of BC cells. Moreover, the expression of SIG1R protein in these groups decreased significantly compared to groups treated with BD1047 or low-dose CdCl2 alone. In conclusion, low-dose CdCl2 is thought to increase the apoptotic ability of BD1047 in BC cells by reducing SIG1R expression.
Collapse
Affiliation(s)
- Barış Yıldız
- Institute of Health Sciences, Department of Physiology, Kafkas University, 36100, Kars, Turkey
| | - Ramazan Demirel
- Department of Bioengineering, Institute of Natural and Applied Sciences, Kafkas University, 36100, Kars, Turkey
| | - Hatice Beşeren Havadar
- Deparment of Medical Pathology, Centre of Health Research and Training Hospital, Kafkas University, 36100, Kars, Turkey
| | - Gülden Yıldız
- Deparment of Medical Pathology, Centre of Health Research and Training Hospital, Kafkas University, 36100, Kars, Turkey
| | - Cem Öziç
- Department of Medical Biology, School of Medicine, Kafkas University, 36100, Kars, Turkey
| | - Nadide Nabil Kamiloğlu
- Department of Physiology, Faculty of Veterinary Medicine, Kafkas University, 36100, Kars, Turkey
| | - Özkan Özden
- Department of Bioengineering, Faculty of Engineering and Architecture, Kafkas University, 36100, Kars, Turkey.
| |
Collapse
|
5
|
Kurihara M, Komatsu H, Sengoku R, Shibukawa M, Morimoto S, Matsubara T, Arakawa A, Orita M, Ishibashi K, Mitsutake A, Shibata S, Ishiura H, Adachi K, Ohse K, Hatano K, Ihara R, Higashihara M, Nishina Y, Tokumaru AM, Ishii K, Saito Y, Murayama S, Kanemaru K, Iwata A. CSF P-Tau181 and Other Biomarkers in Patients With Neuronal Intranuclear Inclusion Disease. Neurology 2023; 100:e1009-e1019. [PMID: 36517236 PMCID: PMC9990848 DOI: 10.1212/wnl.0000000000201647] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/16/2022] [Accepted: 10/11/2022] [Indexed: 12/15/2022] Open
Abstract
BACKGROUND AND OBJECTIVES CSF tau phosphorylated at threonine 181 (p-tau181) is a widely used biomarker for Alzheimer disease (AD) and has recently been regarded to reflect β-amyloid and/or p-tau deposition in the AD brain. Neuronal intranuclear inclusion disease (NIID) is a neurodegenerative disease characterized by intranuclear inclusions in neurons, glial cells, and other somatic cells. Symptoms include dementia, neuropathy, and others. CSF biomarkers were not reported. The objective of this study was to investigate whether CSF biomarkers including p-tau181 are altered in patients with NIID. METHODS This was a retrospective observational study. CSF concentrations of p-tau181, total tau, amyloid-beta 1-42 (Aβ42), monoamine metabolites homovanillic acid (HVA), and 5-hydroxyindole acetic acid (5-HIAA) were compared between 12 patients with NIID, 120 patients with Alzheimer clinical syndrome biologically confirmed based on CSF biomarker profiles, and patients clinically diagnosed with other neurocognitive disorders (dementia with Lewy bodies [DLB], 24; frontotemporal dementia [FTD], 13; progressive supranuclear palsy [PSP], 21; and corticobasal syndrome [CBS], 13). Amyloid PET using Pittsburgh compound B (PiB) was performed in 6 patients with NIID. RESULTS The mean age of patients with NIID, AD, DLB, FTD, PSP, and CBS was 71.3, 74.6, 76.8, 70.2, 75.5, and 71.9 years, respectively. CSF p-tau181 was significantly higher in NIID (72.7 ± 24.8 pg/mL) compared with DLB, PSP, and CBS and was comparable between NIID and AD. CSF p-tau181 was above the cutoff value (50.0 pg/mL) in 11 of 12 patients with NIID (91.7%). Within these patients, only 2 patients showed decreased CSF Aβ42, and these patients showed negative or mild local accumulation in PiB PET, respectively. PiB PET scans were negative in the remaining 4 patients tested. The proportion of patients with increased CSF p-tau181 and normal Aβ42 (A-T+) was significantly higher in NIID (75%) compared with DLB, PSP, and CBS (4.2%, 4.8%, and 7.7%, respectively). CSF HVA and 5-HIAA concentrations were significantly higher in patients with NIID compared with disease controls. DISCUSSION CSF p-tau181 was increased in patients with NIID without amyloid accumulation. Although the deposition of p-tau has not been reported in NIID brains, the molecular mechanism of tau phosphorylation or secretion of p-tau may be altered in NIID.
Collapse
Affiliation(s)
- Masanori Kurihara
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Hiroki Komatsu
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Renpei Sengoku
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Mari Shibukawa
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Satoru Morimoto
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Tomoyasu Matsubara
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Akira Arakawa
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Makoto Orita
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Kenji Ishibashi
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Akihiko Mitsutake
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Shota Shibata
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Hiroyuki Ishiura
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Kaori Adachi
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Kensuke Ohse
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Keiko Hatano
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Ryoko Ihara
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Mana Higashihara
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Yasushi Nishina
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Aya Midori Tokumaru
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Kenji Ishii
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Yuko Saito
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Shigeo Murayama
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Kazutomi Kanemaru
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan
| | - Atsushi Iwata
- From the Department of Neurology (M.K., H.K., R.S., M.S., S.Morimoto., T.M., A.A., K.H., R.I., M.H., Y.N., S.Murayama., K.K., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Neuropathology (the Brain Bank for Aging Research) (R.S., T.M., A.A., M.O., Y.S., S. Murayama), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Department of Neurology (R.S.), The Jikei University School of Medicine, Tokyo; Department of Neurology (M.S.), Toho University Faculty of Medicine, Tokyo; Department of Physiology (S. Morimoto), Keio University School of Medicine, Tokyo; Research Team for Neuroimaging (K. Ishibashi, K. Ishii), Tokyo Metropolitan Institute of Gerontology; Department of Neurology (A.M., S.S., H.I.), Graduate School of Medicine, The University of Tokyo; Research Initiative Center (K.A.), Organization for Research Initiative and Promotion, Tottori University, Yonago; Integrated Research Initiative for Living Well with Dementia (K.O., A.I.), Tokyo Metropolitan Geriatric Hospital and Institution of Gerontology; Department of Diagnostic Radiology (A.M.T.), Tokyo Metropolitan Geriatric Hospital and Institute of Gerontology; Brain Bank for Neurodevelopmental (S. Murayama), Neurological and Psychiatric Disorders, United Graduate School of Child Development, Osaka University, Japan.
| |
Collapse
|
6
|
Lachance V, Bélanger SM, Hay C, Le Corvec V, Banouvong V, Lapalme M, Tarmoun K, Beaucaire G, Lussier MP, Kourrich S. Overview of Sigma-1R Subcellular Specific Biological Functions and Role in Neuroprotection. Int J Mol Sci 2023; 24:1971. [PMID: 36768299 PMCID: PMC9916267 DOI: 10.3390/ijms24031971] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2022] [Revised: 01/15/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
For the past several years, fundamental research on Sigma-1R (S1R) protein has unveiled its necessity for maintaining proper cellular homeostasis through modulation of calcium and lipid exchange between the endoplasmic reticulum (ER) and mitochondria, ER-stress response, and many other mechanisms. Most of these processes, such as ER-stress response and autophagy, have been associated with neuroprotective roles. In fact, improving these mechanisms using S1R agonists was beneficial in several brain disorders including neurodegenerative diseases. In this review, we will examine S1R subcellular localization and describe S1R-associated biological activity within these specific compartments, i.e., the Mitochondrion-Associated ER Membrane (MAM), ER-Lipid Droplet (ER-LD) interface, ER-Plasma Membreane (ER-PM) interface, and the Nuclear Envelope (NE). We also discussed how the dysregulation of these pathways contributes to neurodegenerative diseases, while highlighting the cellular mechanisms and key binding partners engaged in these processes.
Collapse
Affiliation(s)
- Véronik Lachance
- Département des Sciences Biologiques, Université du Québec à Montréal, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3X8, Canada
- Centre d’Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Pavillon des Sciences biologiques, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3Y7, Canada
| | - Sara-Maude Bélanger
- Département des Sciences Biologiques, Université du Québec à Montréal, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3X8, Canada
- Centre d’Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Pavillon des Sciences biologiques, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3Y7, Canada
| | - Célia Hay
- Département des Sciences Biologiques, Université du Québec à Montréal, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3X8, Canada
- Centre d’Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Pavillon des Sciences biologiques, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3Y7, Canada
| | - Victoria Le Corvec
- Département des Sciences Biologiques, Université du Québec à Montréal, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3X8, Canada
- Centre d’Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Pavillon des Sciences biologiques, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3Y7, Canada
| | - Vina Banouvong
- Département des Sciences Biologiques, Université du Québec à Montréal, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3X8, Canada
- Centre d’Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Pavillon des Sciences biologiques, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3Y7, Canada
| | - Mathieu Lapalme
- Département des Sciences Biologiques, Université du Québec à Montréal, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3X8, Canada
- Centre d’Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Pavillon des Sciences biologiques, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3Y7, Canada
| | - Khadija Tarmoun
- Département des Sciences Biologiques, Université du Québec à Montréal, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3X8, Canada
- Centre d’Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Pavillon des Sciences biologiques, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3Y7, Canada
| | - Guillaume Beaucaire
- Département des Sciences Biologiques, Université du Québec à Montréal, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3X8, Canada
| | - Marc P. Lussier
- Centre d’Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Pavillon des Sciences biologiques, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3Y7, Canada
- Département de Chimie, Université du Québec à Montréal, 2101, Rue Jeanne-Mance, Montréal, QC H2X 2J6, Canada
| | - Saïd Kourrich
- Département des Sciences Biologiques, Université du Québec à Montréal, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3X8, Canada
- Centre d’Excellence en Recherche sur les Maladies Orphelines-Fondation Courtois, Pavillon des Sciences biologiques, 141 Avenue du Président-Kennedy, Montréal, QC H2X 3Y7, Canada
- Center for Studies in Behavioral Neurobiology, Concordia University, Montreal, QC H4B 1R6, Canada
| |
Collapse
|
7
|
Munguia-Galaviz FJ, Miranda-Diaz AG, Cardenas-Sosa MA, Echavarria R. Sigma-1 Receptor Signaling: In Search of New Therapeutic Alternatives for Cardiovascular and Renal Diseases. Int J Mol Sci 2023; 24:ijms24031997. [PMID: 36768323 PMCID: PMC9916216 DOI: 10.3390/ijms24031997] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Cardiovascular and renal diseases are among the leading causes of death worldwide, and regardless of current efforts, there is a demanding need for therapeutic alternatives to reduce their progression to advanced stages. The stress caused by diseases leads to the activation of protective mechanisms in the cell, including chaperone proteins. The Sigma-1 receptor (Sig-1R) is a ligand-operated chaperone protein that modulates signal transduction during cellular stress processes. Sig-1R interacts with various ligands and proteins to elicit distinct cellular responses, thus, making it a potential target for pharmacological modulation. Furthermore, Sig-1R ligands activate signaling pathways that promote cardioprotection, ameliorate ischemic injury, and drive myofibroblast activation and fibrosis. The role of Sig-1R in diseases has also made it a point of interest in developing clinical trials for pain, neurodegeneration, ischemic stroke, depression in patients with heart failure, and COVID-19. Sig-1R ligands in preclinical models have significantly beneficial effects associated with improved cardiac function, ventricular remodeling, hypertrophy reduction, and, in the kidney, reduced ischemic damage. These basic discoveries could inform clinical trials for heart failure (HF), myocardial hypertrophy, acute kidney injury (AKI), and chronic kidney disease (CKD). Here, we review Sig-1R signaling pathways and the evidence of Sig-1R modulation in preclinical cardiac and renal injury models to support the potential therapeutic use of Sig-1R agonists and antagonists in these diseases.
Collapse
Affiliation(s)
- Francisco Javier Munguia-Galaviz
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Division de Ciencias de la Salud, Centro Universitario del Sur, Universidad de Guadalajara, Ciudad Guzman 49000, Jalisco, Mexico
| | - Alejandra Guillermina Miranda-Diaz
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Miguel Alejandro Cardenas-Sosa
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Raquel Echavarria
- CONACYT-Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Jalisco, Mexico
- Correspondence:
| |
Collapse
|
8
|
Lenoir S, Lahaye RA, Vitet H, Scaramuzzino C, Virlogeux A, Capellano L, Genoux A, Gershoni-Emek N, Geva M, Hayden MR, Saudou F. Pridopidine rescues BDNF/TrkB trafficking dynamics and synapse homeostasis in a Huntington disease brain-on-a-chip model. Neurobiol Dis 2022; 173:105857. [PMID: 36075537 DOI: 10.1016/j.nbd.2022.105857] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2021] [Revised: 08/30/2022] [Accepted: 09/02/2022] [Indexed: 10/14/2022] Open
Abstract
Huntington disease (HD) is a neurodegenerative disorder caused by polyglutamine-encoding CAG repeat expansion in the huntingtin (HTT) gene. HTT is involved in the axonal transport of vesicles containing brain-derived neurotrophic factor (BDNF). In HD, diminished BDNF transport leads to reduced BDNF delivery to the striatum, contributing to striatal and cortical neuronal death. Pridopidine is a selective and potent sigma-1 receptor (S1R) agonist currently in clinical development for HD. The S1R is located at the endoplasmic reticulum (ER)-mitochondria interface, where it regulates key cellular pathways commonly impaired in neurodegenerative diseases. We used a microfluidic device that reconstitutes the corticostriatal network, allowing the investigation of presynaptic dynamics, synaptic morphology and transmission, and postsynaptic signaling. Culturing primary neurons from the HD mouse model HdhCAG140/+ provides a "disease-on-a-chip" platform ideal for investigating pathogenic mechanisms and drug activity. Pridopidine rescued the trafficking of BDNF and TrkB resulting in an increased neurotrophin signaling at the synapse. This increased the capacity of HD neurons to release glutamate and restored homeostasis at the corticostriatal synapse. These data suggest that pridopidine enhances the availability of corticostriatal BDNF via S1R activation, leading to neuroprotective effects.
Collapse
Affiliation(s)
- Sophie Lenoir
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | - Romane A Lahaye
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | - Hélène Vitet
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | - Chiara Scaramuzzino
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | - Amandine Virlogeux
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | - Laetitia Capellano
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | - Aurélie Genoux
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France
| | | | | | - Michael R Hayden
- Prilenia Therapeutics, Herzliya, Israel; The Centre for Molecular Medicine and Therapeutics, BC Children's Hospital Research Institute, University of British Columbia, Vancouver, British Columbia, Canada
| | - Frédéric Saudou
- Univ. Grenoble Alpes, Inserm, U1216, CHU Grenoble Alpes, Grenoble Institut Neuroscience, GIN, Grenoble, France..
| |
Collapse
|
9
|
Mishiro K, Wang M, Hirata S, Fuchigami T, Shiba K, Kinuya S, Ogawa K. Development of tumor-targeting aza-vesamicol derivatives with high affinity for sigma receptors for cancer theranostics. RSC Med Chem 2022; 13:986-997. [PMID: 36092143 PMCID: PMC9384704 DOI: 10.1039/d2md00099g] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2022] [Accepted: 06/28/2022] [Indexed: 07/31/2023] Open
Abstract
As sigma receptors are highly expressed on various cancer cells, radiolabeled sigma receptor ligands have been developed as imaging and therapeutic probes for cancer. Previously, we synthesized and evaluated a radioiodinated vesamicol derivative, 2-(4-[125I](4-iodophenyl)piperidine)cyclohexanol ((+)-[125I]pIV), and a radioiodinated aza-vesamicol derivative, trans-2-(4-(3-[125I](4-iodophenyl)propyl)piperazin-1-yl)cyclohexan-1-ol ([125I]2), as sigma-1 receptor-targeting probes. In order to obtain sigma receptor-targeting probes with superior biodistribution characteristics, we firstly synthesized twelve bromine-containing aza-vesamicol derivatives and evaluated their affinity for sigma receptors. One such derivative exhibited high selectivity for the sigma-1 receptor and another exhibited high affinity for both the sigma-1 and sigma-2 receptors. Thus, their halogen-substituted iodine- and radioiodine-containing compounds were prepared. The 125I-labeled compounds exhibited high uptake in tumor and lower uptake in non-target tissues than the two previously developed and evaluated 125I-labeled sigma receptor-targeting probes, [125I]pIV and [125I]2. Therefore, these novel radioiodine-labeled compounds should be promising as sigma receptor-targeting probes.
Collapse
Affiliation(s)
- Kenji Mishiro
- Institute for Frontier Science Initiative, Kanazawa University Kakuma-machi Kanazawa Ishikawa 920-1192 Japan
| | - Mengfei Wang
- Graduate School of Pharmaceutical Sciences, Kanazawa University Kakuma-machi Kanazawa Ishikawa 920-1192 Japan
| | - Saki Hirata
- Graduate School of Pharmaceutical Sciences, Kanazawa University Kakuma-machi Kanazawa Ishikawa 920-1192 Japan
| | - Takeshi Fuchigami
- Graduate School of Pharmaceutical Sciences, Kanazawa University Kakuma-machi Kanazawa Ishikawa 920-1192 Japan
| | - Kazuhiro Shiba
- Research Center for Experimental Modeling of Human Disease, Kanazawa University Takara-machi Kanazawa Ishikawa 920-8640 Japan
| | - Seigo Kinuya
- Department of Nuclear Medicine, Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University Takara-machi Kanazawa Ishikawa 920-8641 Japan
| | - Kazuma Ogawa
- Institute for Frontier Science Initiative, Kanazawa University Kakuma-machi Kanazawa Ishikawa 920-1192 Japan
- Graduate School of Pharmaceutical Sciences, Kanazawa University Kakuma-machi Kanazawa Ishikawa 920-1192 Japan
| |
Collapse
|
10
|
Miki Y, Kamata K, Goto S, Sakuraba H, Mori F, Yamagata K, Kijima H, Fukuda S, Wakabayashi K. The clinical and neuropathological picture of adult neuronal intranuclear inclusion disease with no radiological abnormality. Neuropathology 2022; 42:204-211. [PMID: 35274390 DOI: 10.1111/neup.12792] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2021] [Revised: 09/29/2021] [Accepted: 11/02/2021] [Indexed: 11/30/2022]
Abstract
In typical adult neuronal intranuclear inclusion disease (NIID) with predilection for the basal ganglia or cerebral cortex, not only neurons but also glial cells harbor intranuclear inclusions. In addition, these inclusions are present in the peripheral autonomic nervous system, visceral organs and skin. In NIID cases with an expansion of GGC repeats in the 5'-untranslated region (5'-UTR) of the Notch 2 N-terminal like C (NOTCH2NLC) gene, these repeats are located in an upstream open reading frame (uN2C) and result in the production of a polyglycine-containing protein called uN2CpolyG. Typically, patients with adult NIID show high-intensity signals at the corticomedullary junction on diffusion-weighted brain magnetic resonance imaging. We report a case of adult NIID in a 78-year-old Japanese male, who suffered from mild, non-progressive tremor during life but showed no radiographic abnormalities suggestive of adult NIID. Pathologically, ubiquitin-, p62- and uN2CpolyG-positive neuronal intranuclear inclusions were particularly frequent in the hippocampal formation, but were also seen in the enteric plexuses, kidney and cardiac muscles. By contrast, glial intranuclear inclusions were barely evident in the affected regions. The present case also had an immunohistochemical profile differing from that of typical adult NIID. The findings in this case suggest that adult NIID can show clinical, radiographic and pathological heterogeneity.
Collapse
Affiliation(s)
- Yasuo Miki
- Department of Neuropathology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kosuke Kamata
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Shintaro Goto
- Department of Pathology and Bioscience, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Hirotake Sakuraba
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Fumiaki Mori
- Department of Neuropathology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Kazufumi Yamagata
- Department of Bioscience and Laboratory Medicine, Hirosaki University Graduate School of Health Sciences, Hirosaki, Japan
| | - Hiroshi Kijima
- Department of Pathology and Bioscience, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Shinsaku Fukuda
- Department of Gastroenterology and Hematology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| | - Koichi Wakabayashi
- Department of Neuropathology, Hirosaki University Graduate School of Medicine, Hirosaki, Japan
| |
Collapse
|
11
|
Wu NH, Ye Y, Wan BB, Yu YD, Liu C, Chen QJ. Emerging Benefits: Pathophysiological Functions and Target Drugs of the Sigma-1 Receptor in Neurodegenerative Diseases. Mol Neurobiol 2021; 58:5649-5666. [PMID: 34383254 DOI: 10.1007/s12035-021-02524-5] [Citation(s) in RCA: 25] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2021] [Accepted: 08/05/2021] [Indexed: 02/06/2023]
Abstract
The sigma-1 receptor (Sig-1R) is encoded by the SIGMAR1 gene and is a nonopioid transmembrane receptor located in the mitochondrial-associated endoplasmic reticulum membrane (MAM). It helps to locate endoplasmic reticulum calcium channels, regulates calcium homeostasis, and acts as a molecular chaperone to control cell fate and participate in signal transduction. It plays an important role in protecting neurons through a variety of signaling pathways and participates in the regulation of cognition and motor behavior closely related to neurodegenerative diseases. Based on its neuroprotective effects, Sig-1R has now become a breakthrough target for alleviating Alzheimer's disease and other neurodegenerative diseases. This article reviews the most cutting-edge research on the function of Sig-1R under normal or pathologic conditions and target drugs of the sigma-1 receptor in neurodegenerative diseases.
Collapse
Affiliation(s)
- Ning-Hua Wu
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437000, Hubei, China
- Basic Medical College, Hubei University of Science and Technology, Xianning, 437000, Hubei, China
| | - Yu Ye
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437000, Hubei, China
| | - Bin-Bin Wan
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437000, Hubei, China
| | - Yuan-Dong Yu
- Department of Oncology, Renmin Hospital, Hubei University of Medicine, Shiyan, 442000, Hubei, China
| | - Chao Liu
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437000, Hubei, China.
| | - Qing-Jie Chen
- Hubei Key Laboratory of Diabetes and Angiopathy, Hubei University of Science and Technology, Xianning, 437000, Hubei, China.
| |
Collapse
|
12
|
Piechal A, Jakimiuk A, Mirowska-Guzel D. Sigma receptors and neurological disorders. Pharmacol Rep 2021; 73:1582-1594. [PMID: 34350561 PMCID: PMC8641430 DOI: 10.1007/s43440-021-00310-7] [Citation(s) in RCA: 9] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2021] [Revised: 06/27/2021] [Accepted: 07/09/2021] [Indexed: 11/30/2022]
Abstract
Sigma receptors were identified relatively recently, and their presence has been confirmed in the central nervous system and peripheral organs. Changes in sigma receptor function or expression may be involved in neurological diseases, and thus sigma receptors represent a potential target for treating central nervous system disorders. Many substances that are ligands for sigma receptors are widely used in therapies for neurological disorders. In the present review, we discuss the roles of sigma receptors, especially in the central nervous system disorders, and related therapies.
Collapse
Affiliation(s)
- Agnieszka Piechal
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CePT, Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
- Second Department of Neurology, Institute of Psychiatry and Neurology, Sobieskiego 9, 02-957, Warsaw, Poland
| | - Alicja Jakimiuk
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CePT, Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland
| | - Dagmara Mirowska-Guzel
- Department of Experimental and Clinical Pharmacology, Centre for Preclinical Research and Technology CePT, Medical University of Warsaw, Banacha 1B, 02-097, Warsaw, Poland.
| |
Collapse
|
13
|
Dvorácskó S, Lázár L, Fülöp F, Palkó M, Zalán Z, Penke B, Fülöp L, Tömböly C, Bogár F. Novel High Affinity Sigma-1 Receptor Ligands from Minimal Ensemble Docking-Based Virtual Screening. Int J Mol Sci 2021; 22:8112. [PMID: 34360878 PMCID: PMC8347176 DOI: 10.3390/ijms22158112] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/04/2021] [Revised: 07/23/2021] [Accepted: 07/26/2021] [Indexed: 02/07/2023] Open
Abstract
Sigma-1 receptor (S1R) is an intracellular, multi-functional, ligand operated protein that also acts as a chaperone. It is considered as a pluripotent drug target in several pathologies. The publication of agonist and antagonist bound receptor structures has paved the way for receptor-based in silico drug design. However, recent studies on this subject payed no attention to the structural differences of agonist and antagonist binding. In this work, we have developed a new ensemble docking-based virtual screening protocol utilizing both agonist and antagonist bound S1R structures. This protocol was used to screen our in-house compound library. The S1R binding affinities of the 40 highest ranked compounds were measured in competitive radioligand binding assays and the sigma-2 receptor (S2R) affinities of the best S1R binders were also determined. This way three novel high affinity S1R ligands were identified and one of them exhibited a notable S1R/S2R selectivity.
Collapse
Affiliation(s)
- Szabolcs Dvorácskó
- Biological Research Centre, Institute of Biochemistry, Eötvös Loránd Research Network (ELKH), H-6726 Szeged, Hungary; (S.D.); (C.T.)
| | - László Lázár
- Institute of Pharmaceutical Chemistry, University of Szeged, H-6720 Szeged, Hungary; (L.L.); (F.F.); (M.P.); (Z.Z.)
| | - Ferenc Fülöp
- Institute of Pharmaceutical Chemistry, University of Szeged, H-6720 Szeged, Hungary; (L.L.); (F.F.); (M.P.); (Z.Z.)
| | - Márta Palkó
- Institute of Pharmaceutical Chemistry, University of Szeged, H-6720 Szeged, Hungary; (L.L.); (F.F.); (M.P.); (Z.Z.)
| | - Zita Zalán
- Institute of Pharmaceutical Chemistry, University of Szeged, H-6720 Szeged, Hungary; (L.L.); (F.F.); (M.P.); (Z.Z.)
| | - Botond Penke
- Department of Medical Chemistry, University of Szeged, H-6720 Szeged, Hungary;
| | - Lívia Fülöp
- Department of Medical Chemistry, University of Szeged, H-6720 Szeged, Hungary;
| | - Csaba Tömböly
- Biological Research Centre, Institute of Biochemistry, Eötvös Loránd Research Network (ELKH), H-6726 Szeged, Hungary; (S.D.); (C.T.)
| | - Ferenc Bogár
- Department of Medical Chemistry, University of Szeged, H-6720 Szeged, Hungary;
- MTA-SZTE Biomimetic Systems Research Group, Eötvös Loránd Research Network (ELKH), H-6720 Szeged, Hungary
| |
Collapse
|
14
|
Aishwarya R, Abdullah CS, Morshed M, Remex NS, Bhuiyan MS. Sigmar1's Molecular, Cellular, and Biological Functions in Regulating Cellular Pathophysiology. Front Physiol 2021; 12:705575. [PMID: 34305655 PMCID: PMC8293995 DOI: 10.3389/fphys.2021.705575] [Citation(s) in RCA: 48] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
The Sigma 1 receptor (Sigmar1) is a ubiquitously expressed multifunctional inter-organelle signaling chaperone protein playing a diverse role in cellular survival. Recessive mutation in Sigmar1 have been identified as a causative gene for neuronal and neuromuscular disorder. Since the discovery over 40 years ago, Sigmar1 has been shown to contribute to numerous cellular functions, including ion channel regulation, protein quality control, endoplasmic reticulum-mitochondrial communication, lipid metabolism, mitochondrial function, autophagy activation, and involved in cellular survival. Alterations in Sigmar1’s subcellular localization, expression, and signaling has been implicated in the progression of a wide range of diseases, such as neurodegenerative diseases, ischemic brain injury, cardiovascular diseases, diabetic retinopathy, cancer, and drug addiction. The goal of this review is to summarize the current knowledge of Sigmar1 biology focusing the recent discoveries on Sigmar1’s molecular, cellular, pathophysiological, and biological functions.
Collapse
Affiliation(s)
- Richa Aishwarya
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Mahboob Morshed
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Naznin Sultana Remex
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Md Shenuarin Bhuiyan
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States.,Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| |
Collapse
|
15
|
Agha H, McCurdy CR. In vitro and in vivo sigma 1 receptor imaging studies in different disease states. RSC Med Chem 2021; 12:154-177. [PMID: 34046607 PMCID: PMC8127618 DOI: 10.1039/d0md00186d] [Citation(s) in RCA: 12] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2020] [Accepted: 09/02/2020] [Indexed: 12/11/2022] Open
Abstract
The sigma receptor system has been classified into two distinct subtypes, sigma 1 (σ1R) and sigma 2 (σ2R). Sigma 1 receptors (σ1Rs) are involved in many neurodegenerative diseases and different central nervous system disorders such as Alzheimer's disease, Parkinson's disease, schizophrenia, and drug addiction, and pain. This makes them attractive targets for developing radioligands as tools to gain a better understanding of disease pathophysiology and clinical diagnosis. Over the years, several σ1R radioligands have been developed to image the changes in σ1R distribution and density providing insights into their role in disease development. Moreover, the involvement of both σ1Rs and σ2Rs with cancer make these ligands, especially those that are σ2R selective, great tools for imaging different types of tumors. This review will discuss the principles of molecular imaging using PET and SPECT, known σ1R radioligands and their applications for labelling σ1Rs under different disease conditions. Furthermore, this review will highlight σ1R radioligands that have demonstrated considerable potential as biomarkers, and an opportunity to fulfill the ultimate goal of better healthcare outcomes and improving human health.
Collapse
Affiliation(s)
- Hebaalla Agha
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida Gainesville FL 32610 USA +(352) 273 7705 +1 (352) 294 8691
| | - Christopher R McCurdy
- Department of Medicinal Chemistry, College of Pharmacy, University of Florida Gainesville FL 32610 USA +(352) 273 7705 +1 (352) 294 8691
- UF Translational Drug Development Core, University of Florida Gainesville FL 32610 USA
| |
Collapse
|
16
|
Shigeno T, Kozaka T, Kitamura Y, Ogawa K, Taki J, Kinuya S, Shiba K. In vitro and in vivo evaluation of [ 125/123I]-2-[4-(2-iodophenyl)piperidino]cyclopentanol([ 125/123I]-OI5V) as a potential sigma-1 receptor ligand for SPECT. Ann Nucl Med 2021; 35:167-175. [PMID: 33417152 DOI: 10.1007/s12149-020-01552-w] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2020] [Accepted: 10/28/2020] [Indexed: 11/26/2022]
Abstract
INTRODUCTION We investigated the characteristics of radio-iodinated 2-[4-(2-iodophenyl)piperidino]cyclopentanol (OI5V) as a single photon emission computed tomography (SPECT) ligand for mapping sigma-1 receptor (σ-1R), which plays an important role in stress remission in many organs. METHODS OI5V was synthesized from o-bromobenzaldehyde in three steps. OI5V was evaluated for its affinity to VAChT, σ-1 and σ-2 receptor by in vitro competitive binding assays using rat tissues and radioligands, [3H]vesamicol, ( +)-[3H]pentazocine and [3H]DTG, respectively. [125/123I]OI5V was prepared from o-trimethylstannyl-cyclopentanevesamicol (OT5V) by the iododestannylation reaction under no-carrier-added conditions. In vivo biodistribution study of [125I]OI5V in blood, brain regions and major organs of rats was performed at 2, 10, 30 and 60 min post-injection. In vivo blocking study and ex vivo autoradiography were performed to assess the binding selectivity of [125I]OI5V for σ-1 receptor. SPECT-CT imaging study was performed using [123I]OI5V. RESULTS OI5V demonstrated high selective binding affinity for σ-1R in vitro. In the biodistribution study, the blood-brain barrier (BBB) permeability of [125I]OI5V was high and the accumulation of [125I]OI5V in the rat cortex at 2 min post-injection exceeded 2.00%ID/g. In the in vivo blocking study, the accumulation of [125I]OI5V in the brain was significantly blocked by co-administration of 0.5 μmol of SA4503 and 1.0 μmol of pentazocine. Ex vivo autoradiography revealed that the regional brain accumulation of [125I]OI5V was similar to σ-1R-rich regions of the rat brain. SPECT images of [123I]OI5V in the rat brain reflected the distribution of sigma receptors in the brain. CONCLUSIONS This study confirmed that [125/123I]OI5V selectively binds σ-1R in the rat brain in vivo. [123I]OI5V was suggested to be useful as a σ-1R ligand for SPECT.
Collapse
Affiliation(s)
- Taiki Shigeno
- Division of Tracer Kinetics, Advanced Science Research Center, Kanazawa, Ishikawa, Japan
| | - Takashi Kozaka
- Division of Tracer Kinetics, Advanced Science Research Center, Kanazawa, Ishikawa, Japan
- Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa , Ishikawa, Japan
| | - Yoji Kitamura
- Division of Tracer Kinetics, Advanced Science Research Center, Kanazawa, Ishikawa, Japan
- Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa , Ishikawa, Japan
| | - Kazuma Ogawa
- Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa , Ishikawa, Japan
| | - Junichi Taki
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Seigo Kinuya
- Division of Tracer Kinetics, Advanced Science Research Center, Kanazawa, Ishikawa, Japan
- Department of Nuclear Medicine, Kanazawa University Hospital, 13-1 Takara-machi, Kanazawa, Ishikawa, 920-8640, Japan
| | - Kazuhiro Shiba
- Division of Tracer Kinetics, Advanced Science Research Center, Kanazawa, Ishikawa, Japan.
- Institute of Medical, Pharmaceutical and Health Sciences, Kanazawa University, Kanazawa , Ishikawa, Japan.
| |
Collapse
|
17
|
Pontisso I, Combettes L. Role of Sigma-1 Receptor in Calcium Modulation: Possible Involvement in Cancer. Genes (Basel) 2021; 12:139. [PMID: 33499031 PMCID: PMC7911422 DOI: 10.3390/genes12020139] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2020] [Revised: 01/15/2021] [Accepted: 01/19/2021] [Indexed: 12/13/2022] Open
Abstract
Ca2+ signaling plays a pivotal role in the control of cellular homeostasis and aberrant regulation of Ca2+ fluxes have a strong impact on cellular functioning. As a consequence of this ubiquitous role, Ca2+ signaling dysregulation is involved in the pathophysiology of multiple diseases including cancer. Indeed, multiple studies have highlighted the role of Ca2+ fluxes in all the steps of cancer progression. In particular, the transfer of Ca2+ at the ER-mitochondrial contact sites, also known as mitochondrial associated membranes (MAMs), has been shown to be crucial for cancer cell survival. One of the proteins enriched at this site is the sigma-1 receptor (S1R), a protein that has been described as a Ca2+-sensitive chaperone that exerts a protective function in cells in various ways, including the modulation of Ca2+ signaling. Interestingly, S1R is overexpressed in many types of cancer even though the exact mechanisms by which it promotes cell survival are not fully elucidated. This review summarizes the findings describing the roles of S1R in the control of Ca2+ signaling and its involvement in cancer progression.
Collapse
Affiliation(s)
- Ilaria Pontisso
- UMR 1282, INSERM, Laboratoire de Biologie et Pharmacologie Appliquée, Ecole Normale Supérieure Paris Saclay, 91190 Gif Sur Yvette, France;
- Faculté des Sciences, Université Paris-Saclay, 91405 Orsay, France
| | - Laurent Combettes
- UMR 1282, INSERM, Laboratoire de Biologie et Pharmacologie Appliquée, Ecole Normale Supérieure Paris Saclay, 91190 Gif Sur Yvette, France;
- Faculté des Sciences, Université Paris-Saclay, 91405 Orsay, France
| |
Collapse
|
18
|
Lee PT, Liévens JC, Wang SM, Chuang JY, Khalil B, Wu HE, Chang WC, Maurice T, Su TP. Sigma-1 receptor chaperones rescue nucleocytoplasmic transport deficit seen in cellular and Drosophila ALS/FTD models. Nat Commun 2020; 11:5580. [PMID: 33149115 PMCID: PMC7642387 DOI: 10.1038/s41467-020-19396-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2019] [Accepted: 10/13/2020] [Indexed: 02/07/2023] Open
Abstract
In a subgroup of patients with amyotrophic lateral sclerosis (ALS)/Frontotemporal dementia (FTD), the (G4C2)-RNA repeat expansion from C9orf72 chromosome binds to the Ran-activating protein (RanGAP) at the nuclear pore, resulting in nucleocytoplasmic transport deficit and accumulation of Ran in the cytosol. Here, we found that the sigma-1 receptor (Sig-1R), a molecular chaperone, reverses the pathological effects of (G4C2)-RNA repeats in cell lines and in Drosophila. The Sig-1R colocalizes with RanGAP and nuclear pore proteins (Nups) and stabilizes the latter. Interestingly, Sig-1Rs directly bind (G4C2)-RNA repeats. Overexpression of Sig-1Rs rescues, whereas the Sig-1R knockout exacerbates, the (G4C2)-RNA repeats-induced aberrant cytoplasmic accumulation of Ran. In Drosophila, Sig-1R (but not the Sig-1R-E102Q mutant) overexpression reverses eye necrosis, climbing deficit, and firing discharge caused by (G4C2)-RNA repeats. These results on a molecular chaperone at the nuclear pore suggest that Sig-1Rs may benefit patients with C9orf72 ALS/FTD by chaperoning the nuclear pore assembly and sponging away deleterious (G4C2)-RNA repeats.
Collapse
Affiliation(s)
- Pin-Tse Lee
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, 333 Cassell Drive, Baltimore, MD, 21224, USA
- The Ph.D Program for Neural Regenerative Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
- Taiwan Biomaterial Company, 6F, No. 26-1, Sec. 2, Shengyi Rd., Zhubei City, Hsin-Chu County, 30261, Taiwan
| | | | - Shao-Ming Wang
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Jian-Ying Chuang
- The Ph.D Program for Neural Regenerative Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Bilal Khalil
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - Hsiang-En Wu
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, 333 Cassell Drive, Baltimore, MD, 21224, USA
| | - Wen-Chang Chang
- The Ph.D Program for Neural Regenerative Medicine, Taipei Medical University, 250 Wuxing Street, Taipei, 11031, Taiwan
| | - Tangui Maurice
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, 333 Cassell Drive, Baltimore, MD, 21224, USA.
| |
Collapse
|
19
|
Yuan Y, Liu Z, Hou X, Li W, Ni J, Huang L, Hu Y, Liu P, Hou X, Xue J, Sun Q, Tian Y, Jiao B, Duan R, Jiang H, Shen L, Tang B, Wang J. Identification of GGC repeat expansion in the NOTCH2NLC gene in amyotrophic lateral sclerosis. Neurology 2020; 95:e3394-e3405. [PMID: 32989102 DOI: 10.1212/wnl.0000000000010945] [Citation(s) in RCA: 62] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2020] [Accepted: 08/03/2020] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To determine whether the GGC repeats in the NOTCH2NLC gene contribute to amyotrophic lateral sclerosis (ALS). METHODS In this study, 545 patients with ALS and 1,305 healthy controls from mainland China were recruited. Several pathogenic mutations in known ALS-causative genes (including C9ORF72 and ATXN2) and polynucleotide repeat expansions in NOP56 and AR genes were excluded. Repeat-primed PCR and GC-rich PCR were performed to determine the GGC repeat size in NOTCH2NLC. Systematic and targeted clinical evaluations and investigations, including skin biopsy and dynamic electrophysiologic studies, were conducted in the genetically affected patients. RESULTS GGC repeat expansion was observed in 4 patients (numbers of repeats 44, 54, 96, and 143), accounting for ≈0.73% (4 of 545) of all patients with ALS. A comparison with 1,305 healthy controls revealed that GGC repeat expansion in NOTCH2NLC was associated with ALS (Fisher exact test, 4 of 545 vs 0 of 1,305, p = 0.007). Compared to patients with the neuronal intranuclear inclusion disease (NIID) muscle weakness-dominant subtype, patients with ALS phenotype carrying the abnormal repeat expansion tended to have a severe phenotype and rapid deterioration. CONCLUSION Our results suggest that ALS is a specific phenotype of NIID or that GGC expansion in NOTCH2NLC is a factor that modifies ALS. These findings may help clarify the pathogenic mechanism of ALS and may expand the known clinical spectrum of NIID.
Collapse
Affiliation(s)
- Yanchun Yuan
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China
| | - Zhen Liu
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China
| | - Xuan Hou
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China
| | - Wanzhen Li
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China.
| | - Jie Ni
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China
| | - Ling Huang
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China
| | - Yiting Hu
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China
| | - Pan Liu
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China
| | - Xiaorong Hou
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China
| | - Jin Xue
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China
| | - Qiying Sun
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China
| | - Yun Tian
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China
| | - Bin Jiao
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China
| | - Ranhui Duan
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China
| | - Hong Jiang
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China
| | - Lu Shen
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China
| | - Beisha Tang
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China
| | - Junling Wang
- From the Department of Neurology (Y.Y., Z.L., X.H., W.L., J.N., Y.H., P.L., X.H., Q.S., Y.T., B.J., H.J., L.S, B.T., J.W.) and National Clinical Research Center for Geriatric Diseases (H.J., L.S, B.T., J.W.), Xiangya Hospital, Department of Neurology (L.H.), the Third Xiangya Hospital, Laboratory of Medical Genetics (J.X., R.D., H.J., L.S, B.T., J.W.), and Key Laboratory of Hunan Province in Neurodegenerative Disorders (J.H., L.S, B.T., J.W.), Central South University, Changsha, Hunan, PR China.
| |
Collapse
|
20
|
Abstract
Neuronal intranuclear inclusion disease (NIID) is a progressive neurodegenerative disease that had been diagnosed by autopsy until recently, but the number of cases has increased since skin biopsy was reported to be useful in 2011. In 2019, the genetical cause of NIID was identified as the extension of the GGC repeat sequence on the NOTCH2NLC gene, and genetic diagnosis became possible. In NIID, there are two groups: a group onset with cognitive dysfunction, and with leukoencephalopathy on head MRI and a high intensity signal at the corticomedurally junction on DWI, and a group with limb weakness. It is necessary to include NIID in the differential diagnosis of leukoencephalopathy and neuropathy, and it is necessary to combine skin biopsy and genetic testing to accurately diagnose of NIID and promote pathological elucidation.
Collapse
Affiliation(s)
- Jun Sone
- Department of Neurology, National Hospital Organization Suzuka National Hospital
| |
Collapse
|
21
|
Kim EJ, Hwang JHL, Gaus SE, Nana AL, Deng J, Brown JA, Spina S, Lee MJ, Ramos EM, Grinberg LT, Kramer JH, Boxer AL, Gorno-Tempini ML, Rosen HJ, Miller BL, Seeley WW. Evidence of corticofugal tau spreading in patients with frontotemporal dementia. Acta Neuropathol 2020; 139:27-43. [PMID: 31542807 DOI: 10.1007/s00401-019-02075-z] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2019] [Revised: 08/10/2019] [Accepted: 08/11/2019] [Indexed: 01/09/2023]
Abstract
Common neurodegenerative diseases feature progressive accumulation of disease-specific protein aggregates in selectively vulnerable brain regions. Increasing experimental evidence suggests that misfolded disease proteins exhibit prion-like properties, including the ability to seed corruptive templating and self-propagation along axons. Direct evidence for transneuronal spread in patients, however, remains limited. To test predictions made by the transneuronal spread hypothesis in human tissues, we asked whether tau deposition within axons of the corticospinal and corticopontine pathways can be predicted based on clinical syndromes and cortical atrophy patterns seen in frontotemporal lobar degeneration (FTLD). Sixteen patients with Pick's disease, 21 with corticobasal degeneration, and 3 with FTLD-MAPT were included, spanning a range of clinical syndromes across the frontotemporal dementia (FTD) spectrum. Cortical involvement was measured using a neurodegeneration score, a tau score, and a composite score based on semiquantitative ratings and complemented by an MRI-based cortical atrophy W-map based on antemortem imaging. Midbrain cerebral peduncle and pontine base descending fibers were divided into three subregions, representing prefrontopontine, corticospinal, and parieto-temporo-occipital fiber pathways. Tau area fraction was calculated in each subregion and related to clinical syndrome and cortical measures. Within each clinical syndrome, there were predicted relationships between cortical atrophy patterns and axonal tau deposition in midbrain cerebral peduncle and pontine base. Between syndromes, contrasting and predictable patterns of brainstem axonal tau deposition emerged, with, for example, greater tau in prefrontopontine fibers in behavioral variant FTD and in corticospinal fibers in corticobasal syndrome. Finally, semiquantitative and quantitative cortical degeneration scores predicted brainstem axonal tau deposition based on anatomical principles. Taken together, these findings provide important human evidence in support of axonal tau spreading in patients with specific forms of tau-related neurodegeneration.
Collapse
Affiliation(s)
- Eun-Joo Kim
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Medical Research Institute, Busan, Republic of Korea
| | - Ji-Hye L Hwang
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Stephanie E Gaus
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Alissa L Nana
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Jersey Deng
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Jesse A Brown
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Salvatore Spina
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Myung Jun Lee
- Department of Neurology, Pusan National University Hospital, Pusan National University School of Medicine and Medical Research Institute, Busan, Republic of Korea
| | - Eliana Marisa Ramos
- Department of Psychiatry, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, CA, USA
| | - Lea T Grinberg
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
- Department of Pathology, University of California, San Francisco, USA
| | - Joel H Kramer
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Adam L Boxer
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Maria Luisa Gorno-Tempini
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Howard J Rosen
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - Bruce L Miller
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA
| | - William W Seeley
- Department of Neurology, Memory and Aging Center, University of California, 675 Nelson Rising Lane, San Francisco, CA, 94158, USA.
- Department of Pathology, University of California, San Francisco, USA.
| |
Collapse
|
22
|
Yamoah A, Tripathi P, Sechi A, Köhler C, Guo H, Chandrasekar A, Nolte KW, Wruck CJ, Katona I, Anink J, Troost D, Aronica E, Steinbusch H, Weis J, Goswami A. Aggregates of RNA Binding Proteins and ER Chaperones Linked to Exosomes in Granulovacuolar Degeneration of the Alzheimer's Disease Brain. J Alzheimers Dis 2020; 75:139-156. [PMID: 32250292 DOI: 10.3233/jad-190722] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Granulovacuolar degeneration (GVD) occurs in Alzheimer's disease (AD) brain due to compromised autophagy. Endoplasmic reticulum (ER) function and RNA binding protein (RBP) homeostasis regulate autophagy. We observed that the ER chaperones Glucose - regulated protein, 78 KDa (GRP78/BiP), Sigma receptor 1 (SigR1), and Vesicle-associated membrane protein associated protein B (VAPB) were elevated in many AD patients' subicular neurons. However, those neurons which were affected by GVD showed lower chaperone levels, and there was only minor co-localization of chaperones with GVD bodies (GVBs), suggesting that neurons lacking sufficient chaperone-mediated proteostasis enter the GVD pathway. Consistent with this notion, granular, incipient pTau aggregates in human AD and pR5 tau transgenic mouse neurons were regularly co-localized with increased chaperone immunoreactivity, whereas neurons with mature neurofibrillary tangles lacked both the chaperone buildup and significant GVD. On the other hand, APP/PS1 (APPswe/PSEN1dE9) transgenic mouse hippocampal neurons that are devoid of pTau accumulation displayed only few GVBs-like vesicles, which were still accompanied by prominent chaperone buildup. Identifying a potential trigger for GVD, we found cytoplasmic accumulations of RBPs including Matrin 3 and FUS as well as stress granules in GVBs of AD patient and pR5 mouse neurons. Interestingly, we observed that GVBs containing aggregated pTau and pTDP-43 were consistently co-localized with the exosomal marker Flotillin 1 in both AD and pR5 mice. In contrast, intraneuronal 82E1-immunoreactive amyloid-β in human AD and APP/PS1 mice only rarely co-localized with Flotillin 1-positive exosomal vesicles. We conclude that altered chaperone-mediated ER protein homeostasis and impaired autophagy manifesting in GVD are linked to both pTau and RBP accumulation and that some GVBs might be targeted to exocytosis.
Collapse
Affiliation(s)
- Alfred Yamoah
- Institute of Neuropathology, RWTH Aachen University Medical School, Aachen, Germany
- EURON - European Graduate School of Neuroscience
| | - Priyanka Tripathi
- Institute of Neuropathology, RWTH Aachen University Medical School, Aachen, Germany
- EURON - European Graduate School of Neuroscience
| | - Antonio Sechi
- Institute of Biomedical Engineering, Department of Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
| | - Christoph Köhler
- Center for Anatomy, Department II, Medical Faculty, University of Cologne, Cologne, Germany
| | - Haihong Guo
- Institute of Neuropathology, RWTH Aachen University Medical School, Aachen, Germany
| | - Akila Chandrasekar
- Institute of Neuropathology, RWTH Aachen University Medical School, Aachen, Germany
| | - Kay Wilhelm Nolte
- Institute of Neuropathology, RWTH Aachen University Medical School, Aachen, Germany
| | - Christoph Jan Wruck
- Institute of Anatomy and Cell Biology, RWTH Aachen University Medical School, Aachen, Germany
| | - Istvan Katona
- Institute of Neuropathology, RWTH Aachen University Medical School, Aachen, Germany
| | - Jasper Anink
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Dirk Troost
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Eleonora Aronica
- Amsterdam UMC, University of Amsterdam, Department of (Neuro) Pathology, Amsterdam Neuroscience, Amsterdam, The Netherlands
| | - Harry Steinbusch
- Department of Psychiatry and Neuropsychology, School for Mental Health and Neuroscience, Maastricht University, Maastricht, The Netherlands
- EURON - European Graduate School of Neuroscience
| | - Joachim Weis
- Institute of Neuropathology, RWTH Aachen University Medical School, Aachen, Germany
| | - Anand Goswami
- Institute of Neuropathology, RWTH Aachen University Medical School, Aachen, Germany
| |
Collapse
|
23
|
Delprat B, Crouzier L, Su TP, Maurice T. At the Crossing of ER Stress and MAMs: A Key Role of Sigma-1 Receptor? ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2020; 1131:699-718. [PMID: 31646531 DOI: 10.1007/978-3-030-12457-1_28] [Citation(s) in RCA: 33] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Calcium exchanges and homeostasis are finely regulated between cellular organelles and in response to physiological signals. Besides ionophores, including voltage-gated Ca2+ channels, ionotropic neurotransmitter receptors, or Store-operated Ca2+ entry, activity of regulatory intracellular proteins finely tune Calcium homeostasis. One of the most intriguing, by its unique nature but also most promising by the therapeutic opportunities it bears, is the sigma-1 receptor (Sig-1R). The Sig-1R is a chaperone protein residing at mitochondria-associated endoplasmic reticulum (ER) membranes (MAMs), where it interacts with several partners involved in ER stress response, or in Ca2+ exchange between the ER and mitochondria. Small molecules have been identified that specifically and selectively activate Sig-1R (Sig-1R agonists or positive modulators) at the cellular level and that also allow effective pharmacological actions in several pre-clinical models of pathologies. The present review will summarize the recent data on the mechanism of action of Sig-1R in regulating Ca2+ exchanges and protein interactions at MAMs and the ER. As MAMs alterations and ER stress now appear as a common track in most neurodegenerative diseases, the intracellular action of Sig-1R will be discussed in the context of the recently reported efficacy of Sig-1R drugs in pathologies like Alzheimer's disease, Parkinson's disease, Huntington's disease, or amyotrophic lateral sclerosis.
Collapse
Affiliation(s)
- Benjamin Delprat
- MMDN, University of Montpellier, EPHE, INSERM, U1198, Montpellier, France.
| | - Lucie Crouzier
- MMDN, University of Montpellier, EPHE, INSERM, U1198, Montpellier, France
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, IRP, NIDA/NIH, Baltimore, MD, USA
| | - Tangui Maurice
- MMDN, University of Montpellier, EPHE, INSERM, U1198, Montpellier, France
| |
Collapse
|
24
|
Ryskamp DA, Korban S, Zhemkov V, Kraskovskaya N, Bezprozvanny I. Neuronal Sigma-1 Receptors: Signaling Functions and Protective Roles in Neurodegenerative Diseases. Front Neurosci 2019; 13:862. [PMID: 31551669 PMCID: PMC6736580 DOI: 10.3389/fnins.2019.00862] [Citation(s) in RCA: 104] [Impact Index Per Article: 17.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2019] [Accepted: 07/31/2019] [Indexed: 12/12/2022] Open
Abstract
Sigma-1 receptor (S1R) is a multi-functional, ligand-operated protein situated in endoplasmic reticulum (ER) membranes and changes in its function and/or expression have been associated with various neurological disorders including amyotrophic lateral sclerosis/frontotemporal dementia, Alzheimer's (AD) and Huntington's diseases (HD). S1R agonists are broadly neuroprotective and this is achieved through a diversity of S1R-mediated signaling functions that are generally pro-survival and anti-apoptotic; yet, relatively little is known regarding the exact mechanisms of receptor functioning at the molecular level. This review summarizes therapeutically relevant mechanisms by which S1R modulates neurophysiology and implements neuroprotective functions in neurodegenerative diseases. These mechanisms are diverse due to the fact that S1R can bind to and modulate a large range of client proteins, including many ion channels in both ER and plasma membranes. We summarize the effect of S1R on its interaction partners and consider some of the cell type- and disease-specific aspects of these actions. Besides direct protein interactions in the endoplasmic reticulum, S1R is likely to function at the cellular/interorganellar level by altering the activity of several plasmalemmal ion channels through control of trafficking, which may help to reduce excitotoxicity. Moreover, S1R is situated in lipid rafts where it binds cholesterol and regulates lipid and protein trafficking and calcium flux at the mitochondrial-associated membrane (MAM) domain. This may have important implications for MAM stability and function in neurodegenerative diseases as well as cellular bioenergetics. We also summarize the structural and biochemical features of S1R proposed to underlie its activity. In conclusion, S1R is incredibly versatile in its ability to foster neuronal homeostasis in the context of several neurodegenerative disorders.
Collapse
Affiliation(s)
- Daniel A. Ryskamp
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX, United States
| | - Svetlana Korban
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic University, Saint Petersburg, Russia
| | - Vladimir Zhemkov
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX, United States
| | - Nina Kraskovskaya
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic University, Saint Petersburg, Russia
| | - Ilya Bezprozvanny
- Department of Physiology, UT Southwestern Medical Center at Dallas, Dallas, TX, United States
- Laboratory of Molecular Neurodegeneration, Peter the Great St. Petersburg State Polytechnic University, Saint Petersburg, Russia
| |
Collapse
|
25
|
Ogawa K, Masuda R, Mishiro K, Wang M, Kozaka T, Shiba K, Kinuya S, Odani A. Syntheses and evaluation of a homologous series of aza-vesamicol as improved radioiodine-labeled probes for sigma-1 receptor imaging. Bioorg Med Chem 2019; 27:1990-1996. [PMID: 30975500 DOI: 10.1016/j.bmc.2019.03.054] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2018] [Revised: 03/27/2019] [Accepted: 03/29/2019] [Indexed: 10/27/2022]
Abstract
Sigma-1 receptor imaging probes for determining the expression levels are desirable for diagnoses of various diseases and companion diagnoses of therapeutic agents targeting the sigma-1 receptor. In this study, we aimed to develop probes with higher affinity for the sigma-1 receptor. For this purpose, we synthesized and evaluated compounds, namely, vesamicol derivatives, in which alkyl chains of varying chain length were introduced between a piperazine ring and a benzene ring. The binding affinity of the vesamicol derivatives for the sigma-1 receptor tended to increase depending on the length of the alkyl chain between the benzene ring and the piperazine ring. The sigma-1 receptor of 2-(4-(3-phenylpropyl)piperazin-1-yl)cyclohexan-1-ol (5) (Ki = 5.8 nM) exhibited the highest binding affinity; therefore, we introduced radioiodine into the benzene ring in 5. The radioiodine labeled probe [125I]2-(4-(3-(4-iodophenyl)propyl)piperazin-1-yl)cyclohexan-1-ol ([125I]10) showed high accumulation in the sigma-1 receptor expressing DU-145 cells both in vitro and in vivo. Co-injection of [125I]10 with an excess level of a sigma receptor ligand, haloperidol, resulted in a significant decrease in the tumor accumulation in vitro and in vivo, indicating sigma receptor-mediated tumor uptake. These results provide useful information for developing sigma-1 receptor imaging probes.
Collapse
Affiliation(s)
- Kazuma Ogawa
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan; Graduate School of medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan.
| | - Ryohei Masuda
- Graduate School of medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Kenji Mishiro
- Institute for Frontier Science Initiative, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Mengfei Wang
- Graduate School of medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Takashi Kozaka
- Advanced Science Research Centre, Kanazawa University, Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Kazuhiro Shiba
- Advanced Science Research Centre, Kanazawa University, Takara-machi, Kanazawa, Ishikawa 920-8640, Japan
| | - Seigo Kinuya
- Graduate School of medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| | - Akira Odani
- Graduate School of medical Sciences, Kanazawa University, Kakuma-machi, Kanazawa, Ishikawa 920-1192, Japan
| |
Collapse
|
26
|
Ogawa K. Development of Diagnostic and Therapeutic Probes with Controlled Pharmacokinetics for Use in Radiotheranostics. Chem Pharm Bull (Tokyo) 2019; 67:897-903. [PMID: 31474726 DOI: 10.1248/cpb.c19-00274] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The word "theranostics," a portmanteau word made by combining "therapeutics" and "diagnostics," refers to a personalized medicine concept. Recently, the word, "radiotheranostics," has also been used in nuclear medicine as a term that refer to the use of radioisotopes for combined imaging and therapy. For radiotheranostics, a diagnostic probe and a corresponding therapeutic probe can be prepared by introducing diagnostic and therapeutic radioisotopes into the same precursor. These diagnostic and therapeutic probes can be designed to show equivalent pharmacokinetics, which is important for radiotheranostics. As imaging can predict the absorbed radiation dose and thus the therapeutic and side effects, radiotheranostics can help achieve the goal of personalized medicine. In this review, I discuss the use of radiolabeled probes targeting bone metastases, sigma-1 receptor, and αVβ3 integrin for radiotheranostics.
Collapse
Affiliation(s)
- Kazuma Ogawa
- Institute for Frontier Science Initiative, Kanazawa University
- Graduate School of Medical Sciences, Kanazawa University
| |
Collapse
|
27
|
Falcon B, Zhang W, Murzin AG, Murshudov G, Garringer HJ, Vidal R, Crowther RA, Ghetti B, Scheres SHW, Goedert M. Structures of filaments from Pick's disease reveal a novel tau protein fold. Nature 2018; 561:137-140. [PMID: 30158706 PMCID: PMC6204212 DOI: 10.1038/s41586-018-0454-y] [Citation(s) in RCA: 575] [Impact Index Per Article: 82.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 07/18/2018] [Indexed: 02/01/2023]
Abstract
The ordered assembly of tau protein into abnormal filamentous inclusions underlies many human neurodegenerative diseases1. Tau assemblies seem to spread through specific neural networks in each disease2, with short filaments having the greatest seeding activity3. The abundance of tau inclusions strongly correlates with disease symptoms4. Six tau isoforms are expressed in the normal adult human brain-three isoforms with four microtubule-binding repeats each (4R tau) and three isoforms that lack the second repeat (3R tau)1. In various diseases, tau filaments can be composed of either 3R or 4R tau, or of both. Tau filaments have distinct cellular and neuroanatomical distributions5, with morphological and biochemical differences suggesting that they may be able to adopt disease-specific molecular conformations6,7. Such conformers may give rise to different neuropathological phenotypes8,9, reminiscent of prion strains10. However, the underlying structures are not known. Using electron cryo-microscopy, we recently reported the structures of tau filaments from patients with Alzheimer's disease, which contain both 3R and 4R tau11. Here we determine the structures of tau filaments from patients with Pick's disease, a neurodegenerative disorder characterized by frontotemporal dementia. The filaments consist of residues Lys254-Phe378 of 3R tau, which are folded differently from the tau filaments in Alzheimer's disease, establishing the existence of conformers of assembled tau. The observed tau fold in the filaments of patients with Pick's disease explains the selective incorporation of 3R tau in Pick bodies, and the differences in phosphorylation relative to the tau filaments of Alzheimer's disease. Our findings show how tau can adopt distinct folds in the human brain in different diseases, an essential step for understanding the formation and propagation of molecular conformers.
Collapse
Affiliation(s)
| | | | | | | | - Holly J Garringer
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | - Ruben Vidal
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | - Bernardino Ghetti
- Department of Pathology and Laboratory Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | |
Collapse
|
28
|
Mori F, Tanji K, Miki Y, Toyoshima Y, Sasaki H, Yoshida M, Kakita A, Takahashi H, Wakabayashi K. Immunohistochemical localization of exoribonucleases (DIS3L2 and XRN1) in intranuclear inclusion body disease. Neurosci Lett 2018; 662:389-394. [DOI: 10.1016/j.neulet.2017.10.061] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2017] [Revised: 10/26/2017] [Accepted: 10/30/2017] [Indexed: 12/27/2022]
|
29
|
Kubickova J, Lencesova L, Csaderova L, Stracina T, Hudecova S, Babula P, Rozborilova E, Novakova M, Krizanova O. Haloperidol Affects Plasticity of Differentiated NG-108 Cells Through σ1R/IP 3R1 Complex. Cell Mol Neurobiol 2018; 38:181-194. [PMID: 28786032 PMCID: PMC5775985 DOI: 10.1007/s10571-017-0524-y] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2016] [Accepted: 07/19/2017] [Indexed: 11/18/2022]
Abstract
Haloperidol is an antipsychotic agent that primarily acts as an antagonist of D2 dopamine receptors. Besides other receptor systems, it targets sigma 1 receptors (σ1Rs) and inositol 1,4,5-trisphosphate receptors (IP3Rs). Aim of this work was to investigate possible changes in IP3Rs and σ1Rs resulting from haloperidol treatment and to propose physiological consequences in differentiated NG-108 cells, i.e., effect on cellular plasticity. Haloperidol treatment resulted in up-regulation of both type 1 IP3Rs (IP3R1s) and σ1Rs at mRNA and protein levels. Haloperidol treatment did not alter expression of other types of IP3Rs. Calcium release from endoplasmic reticulum (ER) mediated by increased amount of IP3R1s elevated cytosolic calcium and generated ER stress. IP3R1s were bound to σ1Rs, and translocation of this complex from ER to nucleus occurred in the group of cells treated with haloperidol, which was followed by increased nuclear calcium levels. Haloperidol-induced changes in cytosolic, reticular, and nuclear calcium levels were similar when specific σ1 blocker -BD 1047- was used. Changes in calcium levels in nucleus, ER, and cytoplasm might be responsible for alterations in cellular plasticity, because length of neurites increased and number of neurites decreased in haloperidol-treated differentiated NG-108 cells.
Collapse
Affiliation(s)
- Jana Kubickova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Lubomira Lencesova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Lucia Csaderova
- Institute of Virology, Biomedical Research Center, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Tibor Stracina
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Sona Hudecova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia
| | - Petr Babula
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Eva Rozborilova
- Clinics of Pneumology and Phthisiology, Jessenius Faculty of Medicine, Martin, Slovakia
| | - Marie Novakova
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic
| | - Olga Krizanova
- Institute of Clinical and Translational Research, Biomedical Research Center, Slovak Academy of Sciences, Dubravska cesta 9, 84505, Bratislava, Slovakia.
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| |
Collapse
|
30
|
Hong J, Wang L, Zhang T, Zhang B, Chen L. Sigma-1 receptor knockout increases α-synuclein aggregation and phosphorylation with loss of dopaminergic neurons in substantia nigra. Neurobiol Aging 2017; 59:171-183. [PMID: 28870519 DOI: 10.1016/j.neurobiolaging.2017.08.007] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/16/2017] [Revised: 06/20/2017] [Accepted: 08/02/2017] [Indexed: 01/17/2023]
Abstract
Sigma-1 receptor (σ1R) is expressed in dopaminergic neurons of substantia nigra. Here, we show that σ1R knockout (σ1R-/-) mice, at age 6-12 months, appeared with age-related loss of dopaminergic neurons and decline of motor coordination. Levels of α-synuclein (αSyn) oligomers and fibrillar αSyn in substantia nigra of σ1R-/- mice were age-dependently increased without the changes in αSyn monomers. The phosphorylation of αSyn monomers or oligomers in dopaminergic neurons was enhanced in σ1R-/- mice. Levels of phosphorylated eIF2a and C/EBP homologous protein expression were elevated in σ1R-/- mice with decline of proteasome activity. Inhibition of endoplasmic reticulum stress by salubrinal recovered the αSyn phosphorylation and proteasome activity and prevented early oligomerization of αSyn in σ1R-/- mice. Rifampicin reduced the late increase of αSyn oligomers in σ1R-/- mice. Rifampicin or salubrinal could reduce the loss of dopaminergic neurons in σ1R-/- mice and improved their motor coordination. The results indicate that the σ1R deficiency through enhanced aggregation and phosphorylation of αSyn causes the loss of dopaminergic neurons leading to the decline of motor coordination.
Collapse
Affiliation(s)
- Juan Hong
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China; Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Ling Wang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Tingting Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Baofeng Zhang
- Department of Physiology, Nanjing Medical University, Nanjing, China
| | - Ling Chen
- State Key Lab of Reproductive Medicine, Nanjing Medical University, Nanjing, China; Department of Physiology, Nanjing Medical University, Nanjing, China.
| |
Collapse
|
31
|
Yang H, Fu Y, Liu X, Shahi PK, Mavlyutov TA, Li J, Yao A, Guo SZW, Pattnaik BR, Guo LW. Role of the sigma-1 receptor chaperone in rod and cone photoreceptor degenerations in a mouse model of retinitis pigmentosa. Mol Neurodegener 2017; 12:68. [PMID: 28927431 PMCID: PMC5606113 DOI: 10.1186/s13024-017-0202-z] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/26/2017] [Accepted: 08/09/2017] [Indexed: 12/24/2022] Open
Abstract
BACKGROUND Retinitis pigmentosa (RP) is the most common inherited retinal degenerative disease yet with no effective treatment available. The sigma-1 receptor (S1R), a ligand-regulated chaperone, emerges as a potential retina-protective therapeutic target. In particular, pharmacological activation of S1R was recently shown to rescue cones in the rd10 mouse, a rod Pde6b mutant that recapitulates the RP pathology of autonomous rod degeneration followed by secondary death of cones. The mechanisms underlying the S1R protection for cones are not understood in detail. METHODS By rearing rd10/S1R-/- and rd10/S1R+/+ mice in dim light to decelerate rapid rod/cone degeneration, we were able to compare their retinal biochemistry, histology and functions throughout postnatal 3-6 weeks (3 W-6 W). RESULTS The receptor-interacting protein kinases (RIP1/RIP3) and their interaction (proximity ligation) dramatically up-regulated after 5 W in rd10/S1R-/- (versus rd10/S1R+/+) retinas, indicative of intensified necroptosis activation, which was accompanied by exacerbated loss of cones. Greater rod loss in rd10/S1R-/- versus rd10/S1R+/+ retinas was evidenced by more cleaved Caspase3 (4 W) and lower rod electro-retinographic a-waves (4 W-6 W), concomitant with reduced LC3-II and CHOP (4 W-6 W), markers of autophagy and endoplasmic reticulum stress response, respectively. However, the opposite occurred at 3 W. CONCLUSION This study reveals previously uncharacterized S1R-associated mechanisms during rd10 photoreceptor degeneration, including S1R's influences on necroptosis and autophagy as well as its biphasic role in rod degeneration upstream of cone death.
Collapse
Affiliation(s)
- Huan Yang
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
| | - Yingmei Fu
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
- Shanghai Key Laboratory of Psychotic Disorders, Shanghai Mental Health Center, Shanghai Jiao Tong University School of Medicine, 600 Wanping Nan Road, Shanghai, 200030 People’s Republic of China
| | - Xinying Liu
- Department of Pediatrics, Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, 1300 University Avenue, SMI 112, Madison, WI 53706 USA
| | - Pawan K. Shahi
- Department of Pediatrics, Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, 1300 University Avenue, SMI 112, Madison, WI 53706 USA
| | - Timur A. Mavlyutov
- Department of Anesthesiology, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
| | - Jun Li
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
- Department of Ophthalmology, the First Hospital of China Medical University, Shenyang, 110001 People’s Republic of China
- Department of Ophthalmology, the 3rd People’s Hospital of Dalian, Dalian, 116033 People’s Republic of China
| | - Annie Yao
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
| | - Steven Z.-W. Guo
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
| | - Bikash R. Pattnaik
- Department of Pediatrics, Department of Ophthalmology and Visual Sciences, University of Wisconsin School of Medicine and Public Health, 1300 University Avenue, SMI 112, Madison, WI 53706 USA
- McPherson Eye Research Institute, University of Wisconsin, Madison, WI 53705 USA
| | - Lian-Wang Guo
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin School of Medicine and Public Health, Madison, WI 53705 USA
- McPherson Eye Research Institute, University of Wisconsin, Madison, WI 53705 USA
- Department of Surgery and Department of Physiology &Cell Biology, the Ohio State University, Columbus, OH 43210 USA
| |
Collapse
|
32
|
Tadić V, Malci A, Goldhammer N, Stubendorff B, Sengupta S, Prell T, Keiner S, Liu J, Guenther M, Frahm C, Witte OW, Grosskreutz J. Sigma 1 receptor activation modifies intracellular calcium exchange in the G93A hSOD1 ALS model. Neuroscience 2017; 359:105-118. [PMID: 28723387 DOI: 10.1016/j.neuroscience.2017.07.012] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Revised: 06/30/2017] [Accepted: 07/07/2017] [Indexed: 10/19/2022]
Abstract
Aberrations in intracellular calcium (Ca2+) have been well established within amyotrophic lateral sclerosis (ALS), a severe motor neuron disease. Intracellular Ca2+ concentration is controlled in part through the endoplasmic reticulum (ER) mitochondria Ca2+ cycle (ERMCC). The ER supplies Ca2+ to the mitochondria at close contacts between the two organelles, i.e. the mitochondria-associated ER membranes (MAMs). The Sigma 1 receptor (Sig1R) is enriched at MAMs, where it acts as an inter-organelle signaling modulator. However, its impact on intracellular Ca2+ at the cellular level remains to be thoroughly investigated. Here, we used cultured embryonic mice spinal neurons to investigate the influence of Sig1R activation on intracellular Ca2+ homeostasis in the presence of G93AhSOD1 (G93A), an established ALS-causing mutation. Sig1R expression was increased in G93A motor neurons relative to non-transgenic (nontg) controls. Furthermore, we demonstrated significantly reduced bradykinin-sensitive intracellular Ca2+ stores in G93A spinal neurons, which were normalized by the Sig1R agonist SA4503. Moreover, SA4503 accelerated cytosolic Ca2+ clearance following a) AMPAR activation by kainate and b) IP3R-mediated ER Ca2+ release following bradykinin stimulation in both genotypes. PRE-084 (another Sig1R agonist) did not exert any significant effects on cytosolic Ca2+. Both Sig1R expression and functionality were altered by the G93A mutation, indicating the centrality of Sig1R in ALS pathology. Here, we showed that intracellular Ca2+ shuttling can be manipulated by Sig1R activation, thus demonstrating the value of using the pharmacological manipulation of Sig1R to understand Ca2+ homeostasis.
Collapse
Affiliation(s)
- Vedrana Tadić
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Ayse Malci
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Nadine Goldhammer
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Beatrice Stubendorff
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Saikata Sengupta
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Tino Prell
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Silke Keiner
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Jingyu Liu
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Madlen Guenther
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Christiane Frahm
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Otto W Witte
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| | - Julian Grosskreutz
- Hans Berger Department of Neurology, Jena University Hospital, Am Klinikum 1, 07747 Jena, Germany.
| |
Collapse
|
33
|
Weng TY, Tsai SYA, Su TP. Roles of sigma-1 receptors on mitochondrial functions relevant to neurodegenerative diseases. J Biomed Sci 2017; 24:74. [PMID: 28917260 PMCID: PMC5603014 DOI: 10.1186/s12929-017-0380-6] [Citation(s) in RCA: 80] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2017] [Accepted: 09/05/2017] [Indexed: 12/20/2022] Open
Abstract
The sigma-1 receptor (Sig-1R) is a chaperone that resides mainly at the mitochondrion-associated endoplasmic reticulum (ER) membrane (called the MAMs) and acts as a dynamic pluripotent modulator in living systems. At the MAM, the Sig-1R is known to play a role in regulating the Ca2+ signaling between ER and mitochondria and in maintaining the structural integrity of the MAM. The MAM serves as bridges between ER and mitochondria regulating multiple functions such as Ca2+ transfer, energy exchange, lipid synthesis and transports, and protein folding that are pivotal to cell survival and defense. Recently, emerging evidences indicate that the MAM is critical in maintaining neuronal homeostasis. Thus, given the specific localization of the Sig-1R at the MAM, we highlight and propose that the direct or indirect regulations of the Sig-1R on mitochondrial functions may relate to neurodegenerative diseases including Alzheimer's disease (AD), Parkinson's disease (PD), Huntington's disease (HD) and amyotrophic lateral sclerosis (ALS). In addition, the promising use of Sig-1R ligands to rescue mitochondrial dysfunction-induced neurodegeneration is addressed.
Collapse
Affiliation(s)
- Tzu-Yu Weng
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, IRP, NIDA/NIH, Triad Bldg. suite 3512, 333 Cassell Drive, Baltimore, MD 21224 USA
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Shang-Yi Anne Tsai
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, IRP, NIDA/NIH, Triad Bldg. suite 3512, 333 Cassell Drive, Baltimore, MD 21224 USA
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Branch, Intramural Research Program, National Institute on Drug Abuse, NIH, DHHS, IRP, NIDA/NIH, Triad Bldg. suite 3512, 333 Cassell Drive, Baltimore, MD 21224 USA
| |
Collapse
|
34
|
Weng TY, Hung DT, Su TP, Tsai SYA. Loss of Sigma-1 Receptor Chaperone Promotes Astrocytosis and Enhances the Nrf2 Antioxidant Defense. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2017; 2017:4582135. [PMID: 28883901 PMCID: PMC5573104 DOI: 10.1155/2017/4582135] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/31/2017] [Revised: 06/09/2017] [Accepted: 06/28/2017] [Indexed: 12/12/2022]
Abstract
Sigma-1 receptor (Sig-1R) functions as a chaperon that interacts with multiple proteins and lipids and is implicated in neurodegenerative and psychiatric diseases. Here, we used Sig-1R KO mice to examine brain expression profiles of astrocytes and ubiquitinated proteins, which are both hallmarks of central nervous system (CNS) pathologies. Our results showed that Sig-1R KO induces increased glial fibrillary acidic protein (GFAP) expression in primary neuron-glia cultures and in the whole brain of fetus mice with concomitantly increased accumulations of ubiquitinated proteins. Astrogliosis was also observed in the neuron-glia culture. Upon proteasome or autophagy inhibitor treatments, the pronounced ubiquitinated proteins were further increased in Sig-1R KO neurons, indicating that the Sig-1R regulates both protein degradation and quality control systems. We found that Nrf2 (nuclear factor erythroid 2-related factor 2), which functions to overcome the stress condition, was enhanced in the Sig-1R KO systems especially when cells were under stressful conditions. Mutation or deficiency of Sig-1Rs has been observed in neurodegenerative models. Our study identifies the critical roles of Sig-1R in CNS homeostasis and supports the idea that functional complementation pathways are triggered in the Sig-1R KO pathology.
Collapse
Affiliation(s)
- Tzu-Yu Weng
- Genomics Research Center, Academia Sinica, Taipei, Taiwan
| | - Denise T. Hung
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, Department of Health and Human Services, National Institutes of Health, Baltimore, MD 21224, USA
| | - Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, Department of Health and Human Services, National Institutes of Health, Baltimore, MD 21224, USA
| | - Shang-Yi A. Tsai
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, Department of Health and Human Services, National Institutes of Health, Baltimore, MD 21224, USA
| |
Collapse
|
35
|
Mavlyutov TA, Yang H, Epstein ML, Ruoho AE, Yang J, Guo LW. APEX2-enhanced electron microscopy distinguishes sigma-1 receptor localization in the nucleoplasmic reticulum. Oncotarget 2017; 8:51317-51330. [PMID: 28881650 PMCID: PMC5584251 DOI: 10.18632/oncotarget.17906] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2017] [Accepted: 05/04/2017] [Indexed: 12/03/2022] Open
Abstract
The sigma-1 receptor (Sig1R) is an endoplasmic reticulum chaperonin that is attracting tremendous interest as a potential anti-neurodegenerative target. While this membrane protein is known to reside in the inner nuclear envelope (NE) and influences transcription, apparent Sig1R presence in the nucleoplasm is often observed, seemingly contradicting its NE localization. We addressed this confounding issue by applying an antibody-free approach of electron microscopy (EM) to define Sig1R nuclear localization. We expressed APEX2 peroxidase fused to Sig1R-GFP in a Sig1R-null NSC34 neuronal cell line generated with CRISPR-Cas9. APEX2-catalyzed gold/silver precipitation markedly improved EM clarity and confirmed an apparent intra-nuclear presence of Sig1R. However, serial sectioning combined with APEX2-enhanced EM revealed that Sig1R actually resided in the nucleoplasmic reticulum (NR), a specialized nuclear compartment formed via NE invagination into the nucleoplasm. NR cross-sections also indicated Sig1R in ring-shaped NR membranes. Thus, this study distinguishes Sig1R in the NR which could otherwise appear localized in the nucleoplasm if detected with low-resolution methods. Our finding is important for uncovering potential Sig1R regulations in the nucleus.
Collapse
Affiliation(s)
- Timur A Mavlyutov
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin, Madison, WI 53705, USA
| | - Huan Yang
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin, Madison, WI 53705, USA
| | - Miles L Epstein
- Department of Neuroscience, University of Wisconsin, Madison, WI 53706, USA
| | - Arnold E Ruoho
- Department of Neuroscience, University of Wisconsin, Madison, WI 53706, USA
| | - Jay Yang
- Department of Anesthesiology, Wisconsin Institute for Medical Research, University of Wisconsin, Madison, WI 53705, USA
| | - Lian-Wang Guo
- Department of Surgery, Wisconsin Institute for Medical Research, University of Wisconsin, Madison, WI 53705, USA.,McPherson Eye Research Institute, University of Wisconsin, Madison, WI 53705, USA.,Department of Surgery, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA.,Department of Physiology and Cell Biology, Davis Heart and Lung Research Institute, The Ohio State University, Columbus, OH 43210, USA
| |
Collapse
|
36
|
De Smet F, Saiz Rubio M, Hompes D, Naus E, De Baets G, Langenberg T, Hipp MS, Houben B, Claes F, Charbonneau S, Delgado Blanco J, Plaisance S, Ramkissoon S, Ramkissoon L, Simons C, van den Brandt P, Weijenberg M, Van England M, Lambrechts S, Amant F, D'Hoore A, Ligon KL, Sagaert X, Schymkowitz J, Rousseau F. Nuclear inclusion bodies of mutant and wild-type p53 in cancer: a hallmark of p53 inactivation and proteostasis remodelling by p53 aggregation. J Pathol 2017; 242:24-38. [PMID: 28035683 DOI: 10.1002/path.4872] [Citation(s) in RCA: 49] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2016] [Revised: 12/20/2016] [Accepted: 12/27/2016] [Indexed: 01/06/2023]
Abstract
Although p53 protein aggregates have been observed in cancer cell lines and tumour tissue, their impact in cancer remains largely unknown. Here, we extensively screened for p53 aggregation phenotypes in tumour biopsies, and identified nuclear inclusion bodies (nIBs) of transcriptionally inactive mutant or wild-type p53 as the most frequent aggregation-like phenotype across six different cancer types. p53-positive nIBs co-stained with nuclear aggregation markers, and shared molecular hallmarks of nIBs commonly found in neurodegenerative disorders. In cell culture, tumour-associated stress was a strong inducer of p53 aggregation and nIB formation. This was most prominent for mutant p53, but could also be observed in wild-type p53 cell lines, for which nIB formation correlated with the loss of p53's transcriptional activity. Importantly, protein aggregation also fuelled the dysregulation of the proteostasis network in the tumour cell by inducing a hyperactivated, oncogenic heat-shock response, to which tumours are commonly addicted, and by overloading the proteasomal degradation system, an observation that was most pronounced for structurally destabilized mutant p53. Patients showing tumours with p53-positive nIBs suffered from a poor clinical outcome, similar to those with loss of p53 expression, and tumour biopsies showed a differential proteostatic expression profile associated with p53-positive nIBs. p53-positive nIBs therefore highlight a malignant state of the tumour that results from the interplay between (1) the functional inactivation of p53 through mutation and/or aggregation, and (2) microenvironmental stress, a combination that catalyses proteostatic dysregulation. This study highlights several unexpected clinical, biological and therapeutically unexplored parallels between cancer and neurodegeneration. Copyright © 2017 Pathological Society of Great Britain and Ireland. Published by John Wiley & Sons, Ltd.
Collapse
Affiliation(s)
- Frederik De Smet
- The Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.,VIB Center for Brain and Disease Research, Leuven, Belgium.,Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA.,The Broad Institute, Cambridge, MA, USA
| | - Mirian Saiz Rubio
- The Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.,VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Daphne Hompes
- Department of Abdominal Surgery, University Hospitals Gasthuisberg, Leuven, Belgium
| | - Evelyne Naus
- The Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.,VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Greet De Baets
- The Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.,VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Tobias Langenberg
- The Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.,VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Mark S Hipp
- Department of Cellular Biochemistry, Max Planck Institute of Biochemistry, Martinsried, Germany
| | - Bert Houben
- The Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.,VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Filip Claes
- The Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.,VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Sarah Charbonneau
- Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Javier Delgado Blanco
- The Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.,VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Stephane Plaisance
- Nucleomics Core, Flanders Institute for Biotechnology (VIB), Leuven, Belgium
| | - Shakti Ramkissoon
- Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA.,Department of Pathology, Division of Neuropathology, Brigham and Women's Hospital and Children's Hospital Boston, Boston, MA, USA.,Department of Pathology, Harvard Medical School, Boston, MA, USA
| | - Lori Ramkissoon
- Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA
| | - Colinda Simons
- Department of Epidemiology - GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Piet van den Brandt
- Department of Epidemiology - GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Matty Weijenberg
- Department of Epidemiology - GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Manon Van England
- Department of Pathology - GROW School for Oncology and Developmental Biology, Maastricht University, Maastricht, The Netherlands
| | - Sandrina Lambrechts
- Department of Obstetrics and Gynaecology, Division of Gynaecological Oncology, University Hospitals Leuven, KU Leuven, Leuven, Belgium
| | - Frederic Amant
- Department of Obstetrics and Gynaecology, Division of Gynaecological Oncology, University Hospitals Leuven, KU Leuven, Leuven, Belgium.,Centre for Gynaecological Oncology Amsterdam, Netherlands Cancer Institute, Amsterdam, The Netherlands
| | - André D'Hoore
- Department of Abdominal Surgery, University Hospitals Gasthuisberg, Leuven, Belgium
| | - Keith L Ligon
- Department of Medical Oncology, Center for Molecular Oncologic Pathology, Dana-Farber Cancer Institute, Boston, MA, USA.,The Broad Institute, Cambridge, MA, USA.,Department of Pathology, Division of Neuropathology, Brigham and Women's Hospital and Children's Hospital Boston, Boston, MA, USA.,Department of Pathology, Harvard Medical School, Boston, MA, USA.,Department of Pathology, Children's Hospital Boston, Boston, MA, USA
| | - Xavier Sagaert
- Translational Cell and Tissue Research, KU Leuven, Leuven, Belgium
| | - Joost Schymkowitz
- The Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.,VIB Center for Brain and Disease Research, Leuven, Belgium
| | - Frederic Rousseau
- The Switch Laboratory, Department of Cellular and Molecular Medicine, KU Leuven, Leuven, Belgium.,VIB Center for Brain and Disease Research, Leuven, Belgium
| |
Collapse
|
37
|
Takahashi-Fujigasaki J, Nakano Y, Uchino A, Murayama S. Adult-onset neuronal intranuclear hyaline inclusion disease is not rare in older adults. Geriatr Gerontol Int 2017; 16 Suppl 1:51-6. [PMID: 27018283 DOI: 10.1111/ggi.12725] [Citation(s) in RCA: 45] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/27/2015] [Indexed: 12/29/2022]
Abstract
Neuronal intranuclear hyaline inclusion disease (NIHID) is a rare neurodegenerative disorder pathologically characterized by localized neuronal loss, and the presence of eosinophilic intranuclear inclusions in neurons and glial cells. NIHID is a heterogeneous disease entity. It is divided into three clinical subgroups: infantile, juvenile and adult forms. Recently, reports of adult-onset cases have increased. Typical adult-onset NIHID consists of cognitive dysfunction with leukoencephalopathy. This type of adult-onset NIHID can be predicted by characteristic magnetic resonance images, high intensity areas on T2-weighted/fluid-attenuated inversion recovery images and persistent high intensity at the corticomedullary junction in diffusion-weighted images. When clinically suspected, the ante-mortem diagnosis can be made by biopsy. In adult-onset NIHID, nuclear inclusions are found more frequently in glial cells, and moderate to severe white matter degeneration is often associated. Although the underlying pathological mechanisms of NIHID are largely unknown, abnormal intranuclear accumulations of proteins and/or dysfunction of protein degradation systems might be related to the pathogenesis. To further clarify the characteristics of this disease entity, biological and pathological analysis of the patients is indispensable. As this disease entity becomes better known, diagnosed cases are expected to increase. Adult-onset NIHID might not be as extremely rare as previously thought.
Collapse
Affiliation(s)
- Junko Takahashi-Fujigasaki
- Department of Neuropathology (Brain Bank for Aging Research), Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Yuta Nakano
- Bioresource Center for Aging Research, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Akiko Uchino
- Department of Neuropathology (Brain Bank for Aging Research), Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Shigeo Murayama
- Department of Neuropathology (Brain Bank for Aging Research), Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.,Bioresource Center for Aging Research, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.,Neurology, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
38
|
Nguyen L, Lucke-Wold BP, Mookerjee S, Kaushal N, Matsumoto RR. Sigma-1 Receptors and Neurodegenerative Diseases: Towards a Hypothesis of Sigma-1 Receptors as Amplifiers of Neurodegeneration and Neuroprotection. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:133-152. [PMID: 28315269 PMCID: PMC5500918 DOI: 10.1007/978-3-319-50174-1_10] [Citation(s) in RCA: 54] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Sigma-1 receptors are molecular chaperones that may act as pathological mediators and targets for novel therapeutic applications in neurodegenerative diseases. Accumulating evidence indicates that sigma-1 ligands can either directly or indirectly modulate multiple neurodegenerative processes, including excitotoxicity, calcium dysregulation, mitochondrial and endoplasmic reticulum dysfunction, inflammation, and astrogliosis. In addition, sigma-1 ligands may act as disease-modifying agents in the treatment for central nervous system (CNS) diseases by promoting the activity of neurotrophic factors and neural plasticity. Here, we summarize their neuroprotective and neurorestorative effects in different animal models of acute brain injury and chronic neurodegenerative diseases, and highlight their potential role in mitigating disease. Notably, current data suggest that sigma-1 receptor dysfunction worsens disease progression, whereas enhancement amplifies pre-existing functional mechanisms of neuroprotection and/or restoration to slow disease progression. Collectively, the data support a model of the sigma-1 receptor as an amplifier of intracellular signaling, and suggest future clinical applications of sigma-1 ligands as part of multi-therapy approaches to treat neurodegenerative diseases.
Collapse
Affiliation(s)
- Linda Nguyen
- Department of Behavioral Medicine and Psychiatry, School of Medicine, West Virginia University, 930 Chestnut Ridge Road, Morgantown, WV, 26506, USA
- Department of Pharmaceutical Sciences, School of Pharmacy, One Medical Center, West Virginia University, Morgantown, WV, 26506, USA
| | - Brandon P Lucke-Wold
- Graduate Program in Neuroscience, School of Medicine, West Virginia University, One Medical Center Drive, Morgantown, WV, 26506, USA
| | - Shona Mookerjee
- College of Pharmacy, Touro University California, 1310 Club Drive, Vallejo, CA, 94592, USA
| | | | - Rae R Matsumoto
- Department of Behavioral Medicine and Psychiatry, School of Medicine, West Virginia University, 930 Chestnut Ridge Road, Morgantown, WV, 26506, USA.
- College of Pharmacy, Touro University California, 1310 Club Drive, Vallejo, CA, 94592, USA.
| |
Collapse
|
39
|
Mancuso R, Navarro X. Sigma-1 Receptor in Motoneuron Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:235-254. [PMID: 28315275 DOI: 10.1007/978-3-319-50174-1_16] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Amyotrophic Lateral Sclerosis (ALS ) is a neurodegenerative disease affecting spinal cord and brain motoneurons , leading to paralysis and early death. Multiple etiopathogenic mechanisms appear to contribute in the development of ALS , including glutamate excitotoxicity, oxidative stress , protein misfolding, mitochondrial defects, impaired axonal transport, inflammation and glial cell alterations. The Sigma-1 receptor is highly expressed in motoneurons of the spinal cord, particularly enriched in the endoplasmic reticulum (ER) at postsynaptic cisternae of cholinergic C-terminals. Several evidences point to participation of Sigma-1R alterations in motoneuron degeneration. Thus, mutations of the transmembrane domain of the Sigma-1R have been described in familial ALS cases. Interestingly, Sigma-1R KO mice display muscle weakness and motoneuron loss. On the other hand, Sigma-1R agonists promote neuroprotection and neurite elongation through activation of protein kinase C on motoneurons in vitro and in vivo after ventral root avulsion. Remarkably, treatment of SOD1 mice, the most usual animal model of ALS , with Sigma-1R agonists resulted in significantly enhanced motoneuron function and preservation, and increased animal survival. Sigma-1R activation also reduced microglial reactivity and increased the glial expression of neurotrophic factors. Two main interconnected mechanisms seem to underlie the effects of Sigma-1R manipulation on motoneurons: modulation of neuronal excitability and regulation of calcium homeostasis. In addition, Sigma-1R also contributes to regulating protein degradation, and reducing oxidative stress. Therefore, the multi-functional nature of the Sigma-1R represents an attractive target for treating aspects of ALS and other motoneuron diseases .
Collapse
Affiliation(s)
- Renzo Mancuso
- Center for Biological Sciences, University of Southampton, Southampton General Hospital, SO16 6YD, Southampton, UK
| | - Xavier Navarro
- Institute of Neurosciences and Department of Cell Biology, Physiology and Immunology, Universitat Autònoma de Barcelona, and Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Bellaterra, Spain.
| |
Collapse
|
40
|
Cai Y, Yang L, Niu F, Liao K, Buch S. Role of Sigma-1 Receptor in Cocaine Abuse and Neurodegenerative Disease. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:163-175. [PMID: 28315271 DOI: 10.1007/978-3-319-50174-1_12] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
Sigma-1 receptors (Sig-1R) are recognized as a unique class of non-G protein-coupled intracellular protein. Sig-1R binds to its ligand such as cocaine , resulting in dissociation of Sig-1R from mitochondrion-associated ER membrane (MAM) to the endoplasmic reticulum (ER), plasma membrane, and nuclear membrane, regulating function of various proteins. Sig-1R has diverse roles in both physiological as well as in pathogenic processes. The disruption of Sig-1R pathways has been implicated as causative mechanism(s) in the development of both neurodegenerative disorders such as Alzheimer disease (AD ), Parkinson disease (PD ), amyotrophic lateral sclerosis (ALS ) and Huntington Disease (HD ) . Additionally, the interaction of cocaine and Sig-1R has more recently been implicated in potentiating the pathogenesis of HIV-associated neurocognitive disorders (HAND) through impairment of blood-brain barrier (BBB), microglial activation and astrogliosis. On the other hand, restoration of Sig-1R homeostasis has been shown to exert neuroprotective effects. In this review, we provide an overview of how Sig-1R plays a role in the pathogenesis of neurodegenerative disorders and cocaine and implications for future development of therapeutic strategies.
Collapse
Affiliation(s)
- Yu Cai
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, DRC 8024, Omaha, NE, 68198-5880, USA
| | - Lu Yang
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, DRC 8024, Omaha, NE, 68198-5880, USA
| | - Fang Niu
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, DRC 8024, Omaha, NE, 68198-5880, USA
| | - Ke Liao
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, DRC 8024, Omaha, NE, 68198-5880, USA
| | - Shilpa Buch
- Department of Pharmacology and Experimental Neuroscience, University of Nebraska Medical Center, 985880 Nebraska Medical Center, DRC 8024, Omaha, NE, 68198-5880, USA.
| |
Collapse
|
41
|
Wong AYC, Hristova E, Ahlskog N, Tasse LA, Ngsee JK, Chudalayandi P, Bergeron R. Aberrant Subcellular Dynamics of Sigma-1 Receptor Mutants Underlying Neuromuscular Diseases. Mol Pharmacol 2016; 90:238-53. [PMID: 27418673 DOI: 10.1124/mol.116.104018] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 07/11/2016] [Indexed: 12/16/2023] Open
Abstract
The sigma-1 receptor (σ-1R) is an endoplasmic reticulum resident chaperone protein involved in a plethora of cellular functions, and whose disruption has been implicated in a wide range of diseases. Genetic analysis has revealed two σ-1R mutants involved in neuromuscular disorders. A point mutation (E102Q) in the ligand-binding domain results in the juvenile form of amyotrophic lateral sclerosis (ALS16), and a 20 amino-acid deletion (Δ31-50) in the putative cytosolic domain leads to a form of distal hereditary motor neuropathy. We investigated the localization and functional properties of these mutants in cell lines using confocal imaging and electrophysiology. The σ-1R mutants exhibited a significant increase in mobility, aberrant localization, and enhanced block of the inwardly rectifying K(+) channel Kir2.1, compared with the wild-type σ-1R. Thus, these σ-1R mutants have different functional properties that could contribute to their disease phenotypes.
Collapse
Affiliation(s)
- Adrian Y C Wong
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Elitza Hristova
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Nina Ahlskog
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Louis-Alexandre Tasse
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Johnny K Ngsee
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Prakash Chudalayandi
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| | - Richard Bergeron
- Neuroscience, Ottawa Hospital Research Institute, Ottawa (A.Y.C.W., E.H., N.A., L.-A.T., J.K.N, P.C., R.B.), and Department of Cellular and Molecular Medicine, University of Ottawa, Ottawa (J.K.N., R.B.), Ontario, Canada
| |
Collapse
|
42
|
Yang H, Hu HY. Sequestration of cellular interacting partners by protein aggregates: implication in a loss-of-function pathology. FEBS J 2016; 283:3705-3717. [PMID: 27016044 DOI: 10.1111/febs.13722] [Citation(s) in RCA: 66] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2015] [Revised: 03/11/2016] [Accepted: 03/24/2016] [Indexed: 01/09/2023]
Abstract
Protein misfolding and aggregation are a hallmark of several neurodegenerative diseases (NDs). However, how protein aggregation leads to cytotoxicity and neurodegeneration is still controversial. Emerging evidence demonstrates that sequestration of cellular-interacting partners by protein aggregates contributes to the pathogenesis of these diseases. Here, we review current research on sequestration of cellular proteins by protein aggregates and its relation to proteinopathies. Based on different interaction modes, we classify these protein sequestrations into four types: protein coaggregation, domain/motif-mediated sequestration, RNA-assisted sequestration, and sequestration of molecular chaperones. Thus, the cellular essential proteins and/or RNA hijacked by protein aggregates may lose their biological functions, consequently resulting in cytotoxicity and neurodegeneration. We have proposed a hijacking model recapitulating the sequestration process and the loss-of-function pathology of ND.
Collapse
Affiliation(s)
- Hui Yang
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China
| | - Hong-Yu Hu
- State Key Laboratory of Molecular Biology, Institute of Biochemistry and Cell Biology, Shanghai Institutes for Biological Sciences, Chinese Academy of Sciences, Shanghai, China.
| |
Collapse
|
43
|
Srivats S, Balasuriya D, Pasche M, Vistal G, Edwardson JM, Taylor CW, Murrell-Lagnado RD. Sigma1 receptors inhibit store-operated Ca2+ entry by attenuating coupling of STIM1 to Orai1. J Cell Biol 2016; 213:65-79. [PMID: 27069021 PMCID: PMC4828687 DOI: 10.1083/jcb.201506022] [Citation(s) in RCA: 69] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 02/24/2016] [Indexed: 11/24/2022] Open
Abstract
Sigma1 receptors (σ1Rs) are expressed widely; they bind diverse ligands, including psychotropic drugs and steroids, regulate many ion channels, and are implicated in cancer and addiction. It is not known how σ1Rs exert such varied effects. We demonstrate that σ1Rs inhibit store-operated Ca(2+)entry (SOCE), a major Ca(2+)influx pathway, and reduce the Ca(2+)content of the intracellular stores. SOCE was inhibited by expression of σ1R or an agonist of σ1R and enhanced by loss of σ1R or an antagonist. Within the endoplasmic reticulum (ER), σ1R associated with STIM1, the ER Ca(2+)sensor that regulates SOCE. This interaction was modulated by σ1R ligands. After depletion of Ca(2+)stores, σ1R accompanied STIM1 to ER-plasma membrane (PM) junctions where STIM1 stimulated opening of the Ca(2+)channel, Orai1. The association of STIM1 with σ1R slowed the recruitment of STIM1 to ER-PM junctions and reduced binding of STIM1 to PM Orai1. We conclude that σ1R attenuates STIM1 coupling to Orai1 and thereby inhibits SOCE.
Collapse
Affiliation(s)
- Shyam Srivats
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, England, UK
| | - Dilshan Balasuriya
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, England, UK
| | - Mathias Pasche
- MRC Laboratory for Molecular Biology, Cambridge CB2 0QH, England, UK
| | - Gerard Vistal
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, England, UK
| | - J Michael Edwardson
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, England, UK
| | - Colin W Taylor
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, England, UK
| | - Ruth D Murrell-Lagnado
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, England, UK Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton BN1 9QG, England, UK
| |
Collapse
|
44
|
Mori F, Tanji K, Miki Y, Toyoshima Y, Yoshida M, Kakita A, Takahashi H, Utsumi J, Sasaki H, Wakabayashi K. G protein-coupled receptor 26 immunoreactivity in intranuclear inclusions associated with polyglutamine and intranuclear inclusion body diseases. Neuropathology 2015; 36:50-5. [DOI: 10.1111/neup.12237] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/21/2015] [Revised: 07/16/2015] [Accepted: 07/16/2015] [Indexed: 01/04/2023]
Affiliation(s)
- Fumiaki Mori
- Department of Neuropathology; Institute of Brain Science, Hirosaki University Graduate School of Medicine; Hirosaki
| | - Kunikazu Tanji
- Department of Neuropathology; Institute of Brain Science, Hirosaki University Graduate School of Medicine; Hirosaki
| | - Yasuo Miki
- Department of Neuropathology; Institute of Brain Science, Hirosaki University Graduate School of Medicine; Hirosaki
| | - Yasuko Toyoshima
- Department of Pathology; Brain Research Institute, University of Niigata; Niigata
| | - Mari Yoshida
- Department of Neuropathology; Aichi Medical University; Nagakute
| | - Akiyoshi Kakita
- Department of Pathological Neuroscience, Center for Bioresource-based Researches; Brain Research Institute, University of Niigata; Niigata
| | - Hitoshi Takahashi
- Department of Pathology; Brain Research Institute, University of Niigata; Niigata
| | - Jun Utsumi
- Department of Neurology; Hokkaido University Graduate School of Medicine; Sapporo Japan
| | - Hidenao Sasaki
- Department of Neurology; Hokkaido University Graduate School of Medicine; Sapporo Japan
| | - Koichi Wakabayashi
- Department of Neuropathology; Institute of Brain Science, Hirosaki University Graduate School of Medicine; Hirosaki
| |
Collapse
|
45
|
Li X, Hu Z, Liu L, Xie Y, Zhan Y, Zi X, Wang J, Wu L, Xia K, Tang B, Zhang R. A SIGMAR1 splice-site mutation causes distal hereditary motor neuropathy. Neurology 2015; 84:2430-7. [PMID: 26078401 DOI: 10.1212/wnl.0000000000001680] [Citation(s) in RCA: 66] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2014] [Accepted: 03/06/2015] [Indexed: 11/15/2022] Open
Abstract
OBJECTIVE To identify the underlying genetic cause in a consanguineous Chinese family segregating distal hereditary motor neuropathy (dHMN) in an autosomal recessive pattern. METHODS We used whole-exome sequencing and homozygosity mapping to detect the genetic variant in 2 affected individuals of the consanguineous Chinese family with dHMN. RNA analysis of peripheral blood leukocytes and immunofluorescence and immunoblotting of stable cell lines were performed to support the pathogenicity of the identified mutation. RESULTS We identified 3 shared novel homozygous variants in 3 shared homozygous regions of the affected individuals. Sequencing of these 3 variants in family members revealed the c.151+1G>T mutation in SIGMAR1 gene, which located in homozygous region spanning approximately 5.3 Mb at chromosome 9p13.1-p13.3, segregated with the dHMN phenotype. The mutation causes an alternative splicing event and generates a transcript variant with an in-frame deletion of 60 base pairs in exon 1 (c.92_151del), and results in an internally shortened protein σ1R(31_50del). The proteasomal inhibitor treatment increased the intracellular amount of σ1R(31_50del) and led to the formation of nuclear aggregates. Stable expressing σ1R(31_50del) induced endoplasmic reticulum stress and enhanced apoptosis. CONCLUSION The homozygous c.151+1G>T mutation in SIGMAR1 caused a novel form of autosomal recessive dHMN in a Chinese consanguineous family. Endoplasmic reticulum stress may have a role in the pathogenesis of dHMN.
Collapse
Affiliation(s)
- Xiaobo Li
- From the Department of Neurology, the Third Xiangya Hospital (X.L., L.L., Y.X., X.Z., R.Z.), State Key Laboratory of Medical Genetics (Z.H., J.W., K.X., B.T.), and Department of Physiology, Xiangya School of Medicine (X.L., L.W.), Central South University, Changsha, PR China
| | - Zhengmao Hu
- From the Department of Neurology, the Third Xiangya Hospital (X.L., L.L., Y.X., X.Z., R.Z.), State Key Laboratory of Medical Genetics (Z.H., J.W., K.X., B.T.), and Department of Physiology, Xiangya School of Medicine (X.L., L.W.), Central South University, Changsha, PR China
| | - Lei Liu
- From the Department of Neurology, the Third Xiangya Hospital (X.L., L.L., Y.X., X.Z., R.Z.), State Key Laboratory of Medical Genetics (Z.H., J.W., K.X., B.T.), and Department of Physiology, Xiangya School of Medicine (X.L., L.W.), Central South University, Changsha, PR China
| | - Yongzhi Xie
- From the Department of Neurology, the Third Xiangya Hospital (X.L., L.L., Y.X., X.Z., R.Z.), State Key Laboratory of Medical Genetics (Z.H., J.W., K.X., B.T.), and Department of Physiology, Xiangya School of Medicine (X.L., L.W.), Central South University, Changsha, PR China
| | - Yajing Zhan
- From the Department of Neurology, the Third Xiangya Hospital (X.L., L.L., Y.X., X.Z., R.Z.), State Key Laboratory of Medical Genetics (Z.H., J.W., K.X., B.T.), and Department of Physiology, Xiangya School of Medicine (X.L., L.W.), Central South University, Changsha, PR China
| | - Xiaohong Zi
- From the Department of Neurology, the Third Xiangya Hospital (X.L., L.L., Y.X., X.Z., R.Z.), State Key Laboratory of Medical Genetics (Z.H., J.W., K.X., B.T.), and Department of Physiology, Xiangya School of Medicine (X.L., L.W.), Central South University, Changsha, PR China
| | - Junling Wang
- From the Department of Neurology, the Third Xiangya Hospital (X.L., L.L., Y.X., X.Z., R.Z.), State Key Laboratory of Medical Genetics (Z.H., J.W., K.X., B.T.), and Department of Physiology, Xiangya School of Medicine (X.L., L.W.), Central South University, Changsha, PR China
| | - Lixiang Wu
- From the Department of Neurology, the Third Xiangya Hospital (X.L., L.L., Y.X., X.Z., R.Z.), State Key Laboratory of Medical Genetics (Z.H., J.W., K.X., B.T.), and Department of Physiology, Xiangya School of Medicine (X.L., L.W.), Central South University, Changsha, PR China
| | - Kun Xia
- From the Department of Neurology, the Third Xiangya Hospital (X.L., L.L., Y.X., X.Z., R.Z.), State Key Laboratory of Medical Genetics (Z.H., J.W., K.X., B.T.), and Department of Physiology, Xiangya School of Medicine (X.L., L.W.), Central South University, Changsha, PR China
| | - Beisha Tang
- From the Department of Neurology, the Third Xiangya Hospital (X.L., L.L., Y.X., X.Z., R.Z.), State Key Laboratory of Medical Genetics (Z.H., J.W., K.X., B.T.), and Department of Physiology, Xiangya School of Medicine (X.L., L.W.), Central South University, Changsha, PR China
| | - Ruxu Zhang
- From the Department of Neurology, the Third Xiangya Hospital (X.L., L.L., Y.X., X.Z., R.Z.), State Key Laboratory of Medical Genetics (Z.H., J.W., K.X., B.T.), and Department of Physiology, Xiangya School of Medicine (X.L., L.W.), Central South University, Changsha, PR China.
| |
Collapse
|
46
|
Abstract
INTRODUCTION Neuropathic pain is difficult to relieve with standard analgesics and tends to be resistant to opioid therapy. Sigma-1 receptors activated during neuropathic injury may sustain pain. Neuropathic injury activates sigma-1 receptors, which results in activation of various kinases, modulates the activity of multiple ion channels, ligand activated ion channels and voltage-gated ion channels; alters monoamine neurotransmission and dampens opioid receptors G-protein activation. Activation of sigma-1 receptors tonically inhibits opioid receptor G-protein activation and thus dampens analgesic responses. Therefore, sigma-1 receptor antagonists are potential analgesics for neuropathic and adjuvants to opioid therapy. AREAS COVERED This article reviews the importance of sigma-1 receptors as pain generators in multiple animal models in order to illustrate both the importance of these unique receptors in pathologic pain and the potential benefits to sigma-1 receptor antagonists as analgesics. EXPERT OPINION Sigma-1 receptor antagonists have a great potential as analgesics for acute neuropathic injury (herpes zoster, acute postoperative pain and chemotherapy induced neuropathy) and may, as an additional benefit, prevent the development of chronic neuropathic pain. Antagonists are potentially effective as adjuvants to opioid therapy when used early to prevent analgesic tolerance. Drug development is complicated by the complexity of sigma-1 receptor pharmacodynamics and its multiple targets, the lack of a specific sigma-1 receptor antagonist, and potential side effects due to on-target toxicities (cognitive impairment, depression).
Collapse
Affiliation(s)
- Mellar P Davis
- Case Western Reserve University, Taussig Cancer Institute, Cleveland Clinic Lerner School of Medicine, Palliative Medicine and Supportive Oncology Services, Division of Solid Tumor, The Cleveland Clinic , 9500 Euclid Ave, Cleveland, OH 44195 , USA
| |
Collapse
|
47
|
Mavlyutov TA, Epstein M, Guo LW. Subcellular localization of the sigma-1 receptor in retinal neurons - an electron microscopy study. Sci Rep 2015; 5:10689. [PMID: 26033680 PMCID: PMC4649997 DOI: 10.1038/srep10689] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Accepted: 04/27/2015] [Indexed: 11/09/2022] Open
Abstract
The Sigma-1 receptor (S1R) is known to play a protective role in the central nervous system including the retina. A major barrier for understanding the underlying mechanism is an ambiguity of S1R subcellular localizations. We thus conducted the first electron microscopy (EM) study of S1R subcellular distribution in the mouse retina. Immuno-EM imaging showed previously under-appreciated S1R presence in photoreceptor cells. Unlike in other cell types in previous reports, in photoreceptor cells S1R was found in the nuclear envelope but not localized in the endoplasmic reticulum (ER), raising a possibility of S1R-mediated modulatory mechanisms different than conventionally thought. While in bipolar cells S1R was detected only in the nuclear envelope, in ganglion cells S1R was identified predominantly in the nuclear envelope and found in the ER as well. A predominant localization of S1R in the nuclear envelope in all three retinal neurons implicates a potential role of S1R in modulating nuclear activities. Moreover, its absence in the plasma membrane and presence in the subsurface ER cisternae that are juxtaposed to the plasma membrane in ganglion cells may lend mechanistic insights generally important for frequently reported S1R modulations of ion channels in neurons.
Collapse
Affiliation(s)
- Timur A. Mavlyutov
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, 5151 Wisconsin Institute for Medical Research, 1111 Highland Ave, Madison, WI 53705, USA
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, 5151 Wisconsin Institute for Medical Research, 1111 Highland Ave, Madison, WI 53705, USA
| | - Miles Epstein
- Department of Neuroscience, University of Wisconsin School of Medicine and Public Health, 41 Bardeen Medical Laboratory, 470 N Charter Street, Madison, WI 53706, USA
| | - Lian-Wang Guo
- Department of Surgery, University of Wisconsin School of Medicine and Public Health, 5151 Wisconsin Institute for Medical Research, 1111 Highland Ave, Madison, WI 53705, USA
- McPherson Eye Research Institute, University of Wisconsin School of Medicine and Public Health, 5151 Wisconsin Institute for Medical Research, 1111 Highland Ave, Madison, WI 53705, USA
| |
Collapse
|
48
|
Oxombre B, Lee-Chang C, Duhamel A, Toussaint M, Giroux M, Donnier-Maréchal M, Carato P, Lefranc D, Zéphir H, Prin L, Melnyk P, Vermersch P. High-affinity σ1 protein agonist reduces clinical and pathological signs of experimental autoimmune encephalomyelitis. Br J Pharmacol 2015; 172:1769-82. [PMID: 25521311 PMCID: PMC4376455 DOI: 10.1111/bph.13037] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2014] [Revised: 10/16/2014] [Accepted: 11/16/2014] [Indexed: 01/04/2023] Open
Abstract
BACKGROUND AND PURPOSE Selective agonists of the sigma-1 receptor (σ1 protein) are generally reported to protect against neuronal damage and modulate oligodendrocyte differentiation. Human and rodent lymphocytes possess saturable, high-affinity binding sites for compounds binding to the σ1 protein and potential immunomodulatory properties have been described for σ1 protein ligands. Experimental autoimmune encephalomyelitis (EAE) is recognized as a valuable model of the inflammatory aspects of multiple sclerosis (MS). Here, we have assessed the role of a σ1 protein agonist, containing the tetrahydroisoquinoline-hydantoin structure, in EAE. EXPERIMENTAL APPROACH EAE was induced in SJL/J female mice by active immunization with myelin proteolipid protein (PLP)139-151 peptide. The σ1 protein agonist was injected i.p. at the time of immunization (day 0). Disease severity was assessed clinically and by histopathological evaluation of the CNS. Phenotyping of B-cell subsets and regulatory T-cells were performed by flow cytometry in spleen and cervical lymph nodes. KEY RESULTS Prophylactic treatment of EAE mice with the σ1 protein agonist prevented mononuclear cell accumulation and demyelination in brain and spinal cord and increased T2 B-cells and regulatory T-cells, resulting in an overall reduction in the clinical progression of EAE. CONCLUSIONS AND IMPLICATIONS This σ1 protein agonist, containing the tetrahydroisoquinoline-hydantoin structure, decreased the magnitude of inflammation in EAE. This effect was associated with increased proportions of B-cell subsets and regulatory T-cells with potential immunoregulatory functions. Targeting of the σ1 protein might thus provide new therapeutic opportunities in MS.
Collapse
MESH Headings
- Animals
- B-Lymphocytes/immunology
- Brain/drug effects
- Brain/pathology
- Cytokines/blood
- Encephalomyelitis, Autoimmune, Experimental/blood
- Encephalomyelitis, Autoimmune, Experimental/drug therapy
- Encephalomyelitis, Autoimmune, Experimental/immunology
- Encephalomyelitis, Autoimmune, Experimental/pathology
- Female
- Immunoglobulin G/blood
- Lymph Nodes/drug effects
- Lymph Nodes/immunology
- Mice
- Multiple Sclerosis/blood
- Multiple Sclerosis/drug therapy
- Multiple Sclerosis/immunology
- Multiple Sclerosis/pathology
- Myelin Proteolipid Protein/immunology
- Neuroprotective Agents/pharmacology
- Neuroprotective Agents/therapeutic use
- Peptide Fragments/immunology
- Receptors, sigma/agonists
- Spinal Cord/drug effects
- Spinal Cord/pathology
- Spleen/drug effects
- Spleen/immunology
- T-Lymphocytes, Regulatory/immunology
- Sigma-1 Receptor
Collapse
Affiliation(s)
- B Oxombre
- Université de LilleLille, France
- UDSL, EA2686-LIRIC, UFR MédecineLille, France
| | - C Lee-Chang
- Université de LilleLille, France
- UDSL, EA2686-LIRIC, UFR MédecineLille, France
| | - A Duhamel
- Université de LilleLille, France
- UDSL, EA 2694, UFR MédecineLille, France
| | - M Toussaint
- Université de LilleLille, France
- CNRS UMR8161Lille, France
| | - M Giroux
- Université de LilleLille, France
- UDSL, EA2686-LIRIC, UFR MédecineLille, France
- Centre Hospitalier Régional et Universitaire de Lille, Pôle de neurologie–Service de Neurologie DLille, France
| | - M Donnier-Maréchal
- Université de LilleLille, France
- UDSL, EA 4481, UFR PharmacieLille, France
| | - P Carato
- Université de LilleLille, France
- UDSL, EA 4481, UFR PharmacieLille, France
| | - D Lefranc
- Université de LilleLille, France
- UDSL, EA2686-LIRIC, UFR MédecineLille, France
| | - H Zéphir
- Université de LilleLille, France
- UDSL, EA2686-LIRIC, UFR MédecineLille, France
- Centre Hospitalier Régional et Universitaire de Lille, Pôle de neurologie–Service de Neurologie DLille, France
| | - L Prin
- Université de LilleLille, France
- UDSL, EA2686-LIRIC, UFR MédecineLille, France
- Centre Hospitalier Régional et Universitaire de Lille, Pôle d'immunologie–Centre de Biologie Pathologie et GénétiqueLille, France
| | - P Melnyk
- Université de LilleLille, France
- CNRS UMR8161Lille, France
- UDSL, EA 4481, UFR PharmacieLille, France
- Inserm UMR-S1172, Jean-Pierre Aubert Research CenterLille, France
| | - P Vermersch
- Université de LilleLille, France
- UDSL, EA2686-LIRIC, UFR MédecineLille, France
- UDSL, EA 4481, UFR PharmacieLille, France
| |
Collapse
|
49
|
Miki Y, Tanji K, Mori F, Wakabayashi K. Sigma-1 receptor is involved in degradation of intranuclear inclusions in a cellular model of Huntington's disease. Neurobiol Dis 2015; 74:25-31. [PMID: 25449906 DOI: 10.1016/j.nbd.2014.11.005] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2014] [Accepted: 11/04/2014] [Indexed: 02/07/2023] Open
Abstract
The sigma-1 receptor (SIGMAR1) is one of the endoplasmic reticulum (ER) chaperones, which participate in the degradation of misfolded proteins via the ER-related degradation machinery linked to the ubiquitin-proteasome pathway. ER dysfunction in the formation of inclusion bodies in various neurodegenerative diseases has also become evident. Recently, we demonstrated that accumulation of SIGMAR1 was common to neuronal nuclear inclusions in polyglutamine diseases including Huntington's disease. Our study also indicated that SIGMAR1 might shuttle between the cytoplasm and the nucleus. In the present study, we investigated the role of SIGMAR1 in nuclear inclusion (NI) formation, using HeLa cells transfected with N-terminal mutant huntingtin. Cell harboring the mutant huntingtin produced SIGMAR1-positive NIs. SIGMAR1 siRNA and a specific inhibitor of the proteasome (epoxomicin) caused significant accumulation of aggregates in the cytoplasm and nucleus. A specific inhibitor of exportin 1 (leptomycin B) also caused NIs. Huntingtin became insolubilized in Western blot analysis after treatments with SIGMAR1 siRNA and epoxomicin. Furthermore, proteasome activity increased chronologically along with the accumulation of mutant huntingtin, but was significantly reduced in cells transfected with SIGMAR1 siRNA. By contrast, overexpression of SIGMAR1 reduced the accumulation of NIs containing mutant huntingtin. Although the LC3-I level was decreased in cells treated with both SIGMAR1 siRNA and control siRNA, the levels of LC3-II and p62 were unchanged. SIGMAR1 agonist and antagonist had no effect on cellular viability and proteasome activity. These findings suggest that the ubiquitin-proteasome pathway is implicated in NI formation, and that SIGMAR1 degrades aberrant proteins in the nucleus via the ER-related degradation machinery. SIGMAR1 might be a promising candidate for therapy of Huntington's disease.
Collapse
Affiliation(s)
- Yasuo Miki
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Japan.
| | - Kunikazu Tanji
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Japan
| | - Fumiaki Mori
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Japan
| | - Koichi Wakabayashi
- Department of Neuropathology, Institute of Brain Science, Hirosaki University Graduate School of Medicine, 5 Zaifu-cho, Hirosaki, Japan
| |
Collapse
|
50
|
Ruscher K, Wieloch T. The involvement of the sigma-1 receptor in neurodegeneration and neurorestoration. J Pharmacol Sci 2015; 127:30-5. [PMID: 25704015 DOI: 10.1016/j.jphs.2014.11.011] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 11/19/2014] [Accepted: 11/27/2014] [Indexed: 02/07/2023] Open
Abstract
The sigma-1 receptor (Sig-1R) is a single 25 kD polypeptide and a chaperone protein immersed in lipid rafts of the endoplasmic reticulum (ER) where it interacts with mitochondria at the mitochondria-associated ER membrane domain (MAM). Upon activation, the Sig-1R binds to the inositol trisphosphate receptor (IP3R), and modulates cellular calcium (Ca(2+)) homeostasis. Also, the activated Sig-1R modulates plasma membrane receptor and ion channel functions, and may regulate cellular excitability. Further, the Sig-1R promotes trafficking of lipids and proteins essential for neurotransmission, cell growth and motility. Activation of the Sig-1R provides neuroprotection and is neurorestorative in cellular and animal models of neurodegenerative diseases and brain ischaemia. Neuroprotection appears to be due to inhibition of cellular Ca(2+) toxicity and/or inflammation, and neurorestoration may include balancing abberant neurotransmission or stimulation of synaptogenesis, thus remodelling brain connectivity. Single nucleotide polymorphisms and mutations of the SIGMAR1 gene worsen outcome in Alzheimer's disease and myotrophic lateral sclerosis supporting a role of Sig-1R in neurodegenerative disease. The combined neuroprotective and neurorestorative actions of the Sig-1R, provide a broad therapeutic time window of Sig-1R agonists. The Sig-1R is therefore a strong therapeutic target for the development of new treatments for neurodegenerative diseases and stroke.
Collapse
Affiliation(s)
- Karsten Ruscher
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, S-22184 Lund, Sweden
| | - Tadeusz Wieloch
- Laboratory for Experimental Brain Research, Division of Neurosurgery, Department of Clinical Sciences, Lund University, BMC A13, S-22184 Lund, Sweden.
| |
Collapse
|