1
views
0
recommends
+1 Recommend
0 collections
    0
    shares
      • Record: found
      • Abstract: found
      • Article: found
      Is Open Access

      Mitochondria modulatory effects of new TSPO ligands in a cellular model of tauopathies

      research-article

      Read this article at

      Bookmark
          There is no author summary for this article yet. Authors can add summaries to their articles on ScienceOpen to make them more accessible to a non-specialist audience.

          Abstract

          Translocator protein 18 kDa (TSPO) is a mitochondrial protein located in the outer membrane and involved in cholesterol translocation, a prerequisite for steroid biosynthesis. TSPO modulation also appears to play a role in other mitochondrial functions, including mitochondrial respiration and cell survival. In the central nervous system, its expression is up‐regulated in neuropathology such as Alzheimer's disease (AD). Previously, we demonstrated that two new TSPO ligands, named 2a and 2b, stimulated pregnenolone synthesis and ATP production in a cellular model of AD overproducing β‐amyloid peptide. The present study aimed to evaluate the impact of the new TSPO ligands on mitochondrial dysfunction in a cellular model of AD‐related tauopathy (human neuroblastoma cells SH‐SY5Y stably overexpressing the P301L‐mutant Tau) presenting mitochondrial impairments, including a decreased ATP synthesis and mitochondrial membrane potential, as well as a decrease in pregnenolone synthesis compared to control cells. The effects of our new ligands were compared with those of TSPO ligands described in the literature (XBD173, SSR‐180,575 and Ro5‐4864). The TSPO ligands 2a and 2b exerted beneficial mitochondrial modulatory effects by increasing ATP levels and mitochondrial membrane potential, paralleled by an increase of pregnenolone levels in mutant Tau cells, as well as in control cells. The compounds 2a and 2b showed effects on mitochondrial activity similar to those obtained with the TSPO ligands of reference. These findings indicate that the new TSPO ligands modulate the mitochondrial bioenergetic phenotype as well as the de novo synthesis of neurosteroids in a cellular model of AD‐related tauopathy, suggesting that these compounds could be potential new therapeutic tools for the treatment of AD.

          Related collections

          Most cited references39

          • Record: found
          • Abstract: found
          • Article: not found

          Allopregnanolone reverses neurogenic and cognitive deficits in mouse model of Alzheimer's disease.

          Our previous analyses showed that allopregnanolone (APalpha) significantly increased proliferation of rodent and human neural progenitor cells in vitro. In this study, we investigated the efficacy of APalpha to promote neurogenesis in the hippocampal subgranular zone (SGZ), to reverse learning and memory deficits in 3-month-old male triple transgenic mouse model of Alzheimer's (3xTgAD) and the correlation between APalpha-induced neural progenitor cell survival and memory function in 3xTgAD mice. Neural progenitor cell proliferation was determined by unbiased stereological analysis of BrdU incorporation and survival determined by FACS for BrdU+ cells. Learning and memory function was assessed using the hippocampal-dependent trace eye-blink conditioning paradigm. At 3 months, basal level of BrdU+ cells in the SGZ of 3xTgAD mice was significantly lower relative to non-Tg mice, despite the lack of evident AD pathology. APalpha significantly increased, in a dose-dependent manner, BrdU+ cells in SGZ in 3xTgAD mice and restored SGZ proliferation to normal magnitude. As with the deficit in proliferation, 3xTgAD mice exhibited deficits in learning and memory. APalpha reversed the cognitive deficits to restore learning and memory performance to the level of normal non-Tg mice. In 3xTgAD mice, APalpha-induced survival of neural progenitors was significantly correlated with APalpha-induced memory performance. These findings suggest that early neurogenic deficits, which were evident before immunodetectable Abeta, may contribute to the cognitive phenotype of AD, and that APalpha could serve as a regenerative therapeutic to prevent or delay neurogenic and cognitive deficits associated with mild cognitive impairment and Alzheimer's disease.
            Bookmark
            • Record: found
            • Abstract: found
            • Article: not found

            The neurosteroid allopregnanolone promotes proliferation of rodent and human neural progenitor cells and regulates cell-cycle gene and protein expression.

            Our previous research demonstrated that the neuroactive progesterone metabolite allopregnanolone (3alpha-hydroxy-5alpha-pregnan-20-one) rapidly induced hippocampal neuron neurite regression (Brinton, 1994). We hypothesized that allopregnanolone-induced neurite regression was a prelude to mitogenesis initiated by a rise in intracellular calcium. Supporting this hypothesis, the current data demonstrate that allopregnanolone, in a dose-dependent manner, induces a significant increase in proliferation of neuroprogenitor cells (NPCs) derived from the rat hippocampus and human neural stem cells (hNSCs) derived from the cerebral cortex. Proliferation was determined by incorporation of bromodeoxyuridine and [3H]thymidine, fluorescence-activated cell sorter analysis of murine leukemia virus-green fluorescent protein-labeled mitotic NPCs, and total cell number counting. Allopregnanolone-induced proliferation was isomer and steroid specific, in that the stereoisomer 3beta-hydroxy-5beta-pregnan-20-one and related steroids did not increase [3H]thymidine uptake. Immunofluorescent analyses for the NPC markers nestin and Tuj1 indicated that newly formed cells were of neuronal lineage. Furthermore, microarray analysis of cell-cycle genes and real-time reverse transcription-PCR and Western blot validation revealed that allopregnanolone increased the expression of genes that promote mitosis and inhibited the expression of genes that repress cell proliferation. Allopregnanolone-induced proliferation was antagonized by the voltage-gated L-type calcium channel (VGLCC) blocker nifedipine, consistent with the finding that allopregnanolone induces a rapid increase in intracellular calcium in hippocampal neurons via a GABA type A receptor-activated VGLCC (Son et al., 2002). These data demonstrate that allopregnanolone significantly increased rat NPC and hNSC proliferation with concomitant regulation in mitotic cell-cycle genes via a VGLCC mechanism. The therapeutic potential of allopregnanolone as a neurogenic molecule is discussed.
              Bookmark
              • Record: found
              • Abstract: found
              • Article: not found

              Ligand for translocator protein reverses pathology in a mouse model of Alzheimer's disease.

              Ligands of the translocator protein (TSPO) elicit pleiotropic neuroprotective effects that represent emerging treatment strategies for several neurodegenerative conditions. To investigate the potential of TSPO as a therapeutic target for Alzheimer's disease (AD), the current study assessed the effects of the TSPO ligand Ro5-4864 on the development of neuropathology in 3xTgAD mice. The effects of the TSPO ligand on neurosteroidogenesis and AD-related neuropathology, including β-amyloid accumulation, gliosis, and behavioral impairment, were examined under both early intervention (7-month-old young-adult male mice with low pathology) and treatment (24-month-old, aged male mice with advanced neuropathology) conditions. Ro5-4864 treatment not only effectively attenuated development of neuropathology and behavioral impairment in young-adult mice but also reversed these indices in aged 3xTgAD mice. Reduced levels of soluble β-amyloid were also observed by the combination of TSPO ligands Ro5-4864 and PK11195 in nontransgenic mice. These findings suggest that TSPO is a promising target for the development of pleiotropic treatment strategies for the management of AD.
                Bookmark

                Author and article information

                Contributors
                anne.eckert@upk.ch
                Journal
                J Neuroendocrinol
                J. Neuroendocrinol
                10.1111/(ISSN)1365-2826
                JNE
                Journal of Neuroendocrinology
                John Wiley and Sons Inc. (Hoboken )
                0953-8194
                1365-2826
                13 October 2019
                January 2020
                : 32
                : 1 , Special Issue of papers from the 10th International Meeting STEROIDS and NERVOUS SYSTEM TORINO, ITALY – February 16th‐20th, 2019 ( doiID: 10.1111/jne.v32.1 )
                : e12796
                Affiliations
                [ 1 ] Transfaculty Research Platform, Molecular & Cognitive Neuroscience Neurobiology Laboratory for Brain Aging and Mental Health University of Basel Basel Switzerland
                [ 2 ] Psychiatric University Clinics Basel Switzerland
                [ 3 ] Laboratoire d’Innovation Thérapeutique Faculté de Pharmacie UMR7200 CNRS Université de Strasbourg Illkirch France
                [ 4 ] Clem Jones Centre for Ageing Dementia Research (CJCADR) Queensland Brain Institute (QBI) The University of Queensland St Lucia QLD Australia
                Author notes
                [*] [* ] Correspondence

                Anne Eckert, Neurobiology Lab for Brain Aging and Mental Health, Transfaculty Research Platform Molecular & Cognitive Neuroscience (MCN), University of Basel, Psychiatric University Clinics Basel Wilhelm Klein‐Strasse 27, Basel CH‐4002, Switzerland.

                Email: anne.eckert@ 123456upk.ch

                Author information
                https://orcid.org/0000-0003-3323-1756
                https://orcid.org/0000-0002-5324-8160
                https://orcid.org/0000-0001-8501-7896
                https://orcid.org/0000-0002-4122-0929
                https://orcid.org/0000-0002-9341-3669
                Article
                JNE12796
                10.1111/jne.12796
                7003898
                31536662
                77a7b7fc-7992-4355-a6ae-7352f9b7f59d
                © 2019 The Authors. Journal of Neuroendocrinology published by John Wiley & Sons Ltd on behalf of British Society for Neuroendocrinology

                This is an open access article under the terms of the http://creativecommons.org/licenses/by-nc/4.0/ License, which permits use, distribution and reproduction in any medium, provided the original work is properly cited and is not used for commercial purposes.

                History
                : 15 April 2019
                : 09 September 2019
                : 17 September 2019
                Page count
                Figures: 7, Tables: 0, Pages: 10, Words: 6617
                Funding
                Funded by: Stiftung Synapsis ‐ Alzheimer Forschung Schweiz AFS , open-funder-registry 10.13039/501100008947;
                Funded by: UPK research Fonds
                Funded by: Schweizerischer Nationalfonds zur Förderung der Wissenschaftlichen Forschung , open-funder-registry 10.13039/501100001711;
                Award ID: #31003A_149728
                Funded by: National Science Foundation , open-funder-registry 10.13039/100000001;
                Categories
                Original Article
                Original Articles
                Custom metadata
                2.0
                January 2020
                Converter:WILEY_ML3GV2_TO_JATSPMC version:5.7.5 mode:remove_FC converted:06.02.2020

                Endocrinology & Diabetes
                alzheimer's disease,bioenergetics,mitochondria,pregnenolone,tau protein,tspo ligands

                Comments

                Comment on this article