1
|
Oh JY, Lee H, Jang SY, Kim H, Park G, Serikov A, Jang JH, Kim J, Yang S, Sa M, Lee SE, Han YE, Hwang TY, Jung SJ, Kim HY, Lee SE, Oh SJ, Kim J, Kim J, Kim J, McHugh TJ, Lee CJ, Nam MH, Park HJ. Central Role of Hypothalamic Circuits for Acupuncture's Anti-Parkinsonian Effects. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2024:e2403245. [PMID: 39119926 DOI: 10.1002/advs.202403245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Revised: 06/30/2024] [Indexed: 08/10/2024]
Abstract
Despite clinical data stretching over millennia, the neurobiological basis of the effectiveness of acupuncture in treating diseases of the central nervous system has remained elusive. Here, using an established model of acupuncture treatment in Parkinson's disease (PD) model mice, we show that peripheral acupuncture stimulation activates hypothalamic melanin-concentrating hormone (MCH) neurons via nerve conduction. We further identify two separate neural pathways originating from anatomically and electrophysiologically distinct MCH neuronal subpopulations, projecting to the substantia nigra and hippocampus, respectively. Through chemogenetic manipulation specifically targeting these MCH projections, their respective roles in mediating the acupuncture-induced motor recovery and memory improvements following PD onset are demonstrated, as well as the underlying mechanisms mediating recovery from dopaminergic neurodegeneration, reactive gliosis, and impaired hippocampal synaptic plasticity. Collectively, these MCH neurons constitute not only a circuit-based explanation for the therapeutic effectiveness of traditional acupuncture, but also a potential cellular target for treating both motor and non-motor PD symptoms.
Collapse
Affiliation(s)
- Ju-Young Oh
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Studies of Translational Acupuncture Research (STAR), Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hyowon Lee
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Sun-Young Jang
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Studies of Translational Acupuncture Research (STAR), Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hyunjin Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of KHU-KIST Convergence Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Geunhong Park
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Almas Serikov
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jae-Hwan Jang
- Studies of Translational Acupuncture Research (STAR), Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Junyeop Kim
- Laboratory of Stem Cells & Cell Reprogramming, Department of Chemistry, Dongguk University, Seoul, 04629, Republic of Korea
| | - Seulkee Yang
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Moonsun Sa
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Sung Eun Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Young-Eun Han
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Tae-Yeon Hwang
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Studies of Translational Acupuncture Research (STAR), Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Sharon Jiyoon Jung
- Technological Convergence Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Hee Young Kim
- Department of Physiology, Yonsei University, Seoul, 03722, Republic of Korea
| | - Seung Eun Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
- Research Animal Resource Center, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Soo-Jin Oh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jeongjin Kim
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
| | - Jeongyeon Kim
- Emotion, Cognition & Behavior Research Group, Korea Brain Research Institute, Daegu, 41062, Republic of Korea
| | - Jongpil Kim
- Laboratory of Stem Cells & Cell Reprogramming, Department of Chemistry, Dongguk University, Seoul, 04629, Republic of Korea
| | - Thomas J McHugh
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Laboratory for Circuit and Behavioral Physiology, RIKEN, Wako-shi Saitama, 351-0198, Japan
| | - C Justin Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Republic of Korea
| | - Min-Ho Nam
- Brain Science Institute, Korea Institute of Science and Technology (KIST), Seoul, 02792, Republic of Korea
- Department of KHU-KIST Convergence Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea
| | - Hi-Joon Park
- College of Korean Medicine, Kyung Hee University, Seoul, 02447, Republic of Korea
- Studies of Translational Acupuncture Research (STAR), Kyung Hee University, Seoul, 02447, Republic of Korea
- Department of KHU-KIST Convergence Science and Technology, Kyung Hee University, Seoul, 02447, Republic of Korea
| |
Collapse
|
2
|
Taraschenko O, Fox HS, Heliso P, Al-Saleem F, Dessain S, Kim WY, Samuelson MM, Dingledine R. Memory loss and aberrant neurogenesis in mice exposed to patient anti-N-methyl-d-aspartate receptor antibodies. Exp Neurol 2024; 378:114838. [PMID: 38801989 DOI: 10.1016/j.expneurol.2024.114838] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2024] [Revised: 05/14/2024] [Accepted: 05/24/2024] [Indexed: 05/29/2024]
Abstract
OBJECTIVE Anti-N-methyl-d-aspartate receptor (anti-NMDAR) encephalitis results in chronic epilepsy and permanent cognitive impairment. One of the possible causes of cognitive impairment in anti-NMDAR could be aberrant neurogenesis, an established contributor to memory loss in idiopathic drug-resistant epilepsy. We developed a mouse model of anti-NMDAR encephalitis and showed that mice exposed to patient anti-NMDAR antibodies for 2 weeks developed seizures and memory loss. In the present study, we assessed the delayed effects of patient-derived antibodies on cognitive phenotype and examined the corresponding changes in hippocampal neurogenesis. METHODS Monoclonal anti-NMDAR antibodies or control antibodies were continuously infused into the lateral ventricle of male C56BL/6J mice (8-12 weeks) via osmotic minipumps for 2 weeks. The motor and anxiety phenotypes were assessed using the open field paradigm, and hippocampal memory and learning were assessed using the object location, Y maze, and Barnes maze paradigms during weeks 1 and 3-4 of antibody washout. The numbers of newly matured granule neurons (Prox-1+) and immature progenitor cells (DCX+) as well as their spatial distribution within the hippocampus were assessed at these time points. Bromodeoxyuridine (BrdU, 50 mg/kg, i.p., daily) was injected on days 2-12 of the infusion, and proliferating cell immunoreactivity was compared in antibody-treated mice and control mice during week 4 of the washout. RESULTS Mice infused with anti-NMDAR antibodies demonstrated spatial memory impairment during week 1 of antibody washout (p = 0.02, t-test; n = 9-11). Histological analysis of hippocampal sections from these mice revealed an increased ectopic displacement of Prox-1+ cells in the dentate hilus compared to the control-antibody-treated mice (p = 0.01; t-test). Mice exposed to anti-NMDAR antibodies also had an impairment of spatial memory and learning during weeks 3-4 of antibody washout (object location: p = 0.009; t-test; Y maze: p = 0.006, t-test; Barnes maze: p = 0.008, ANOVA; n = 8-10). These mice showed increased ratios of the low proliferating (bright) to fast proliferating (faint) BrdU+ cell counts and decreased number of DCX+ cells in the hippocampal dentate gyrus (p = 0.006 and p = 0.04, respectively; t-tests) suggesting ectopic migration and delayed cell proliferation. SIGNIFICANCE These findings suggest that memory and learning impairments induced by patient anti-NMDAR antibodies are sustained upon removal of antibodies and are accompanied by aberrant hippocampal neurogenesis. Interventions directed at the manipulation of neuronal plasticity in patients with encephalitis and cognitive loss may be protective and therapeutically relevant.
Collapse
Affiliation(s)
- Olga Taraschenko
- Department of Neurological Sciences, Division of Epilepsy, University of Nebraska Medical Center, Omaha, NE, United States of America.
| | - Howard S Fox
- Department of Neurological Sciences, Division of Epilepsy, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Priscilla Heliso
- Department of Neurological Sciences, Division of Epilepsy, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Fetweh Al-Saleem
- Lankenau Institute for Medical Research, Wynnewood, PA, United States of America
| | - Scott Dessain
- Lankenau Institute for Medical Research, Wynnewood, PA, United States of America
| | - Woo-Yang Kim
- Department of Biological Sciences, Kent State University, Kent, OH, United States of America
| | - Mystera M Samuelson
- Department of Environmental, Agricultural and Occupational Health, University of Nebraska Medical Center, Omaha, NE, United States of America
| | - Raymond Dingledine
- Department of Pharmacology and Chemical Biology, Emory University School of Medicine, Atlanta, GA, United States of America
| |
Collapse
|
3
|
Crouzier L, Meunier J, Carles A, Morilleau A, Vrigneau C, Schmitt M, Bourguignon JJ, Delprat B, Maurice T. Convolamine, a tropane alkaloid extracted from Convolvulus plauricalis, is a potent sigma-1 receptor-positive modulator with cognitive and neuroprotective properties. Phytother Res 2024; 38:694-712. [PMID: 38011416 DOI: 10.1002/ptr.8068] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 10/26/2023] [Accepted: 10/28/2023] [Indexed: 11/29/2023]
Abstract
BACKGROUND AND AIM By using an in vivo phenotypic screening assay in zebrafish, we identified Convolamine, a tropane alkaloid from Convulvus plauricalis, as a positive modulator of the sigma-1 receptor (S1R). The wfs1abKO zebrafish larva, a model of Wolfram syndrome, exhibits an increased visual-motor response due to a mutation in Wolframin, a protein involved in endoplasmic reticulum-mitochondria communication. We previously reported that ligand activating S1R, restored the cellular and behavioral deficits in patient fibroblasts and zebrafish and mouse models. EXPERIMENTAL PROCEDURES We screened a library of 108 repurposing and natural compounds on zebrafish motor response. KEY RESULTS One hit, the tropane alkaloid Convolamine, restored normal mobility in wfs1abKO larvae without affecting wfs1abWT controls. They did not bind to the S1R agonist/antagonist binding site nor dissociated S1R from BiP, an S1R activity assay in vitro, but behaved as a positive modulator by shifting the IC50 value of the reference agonist PRE-084 to lower values. Convolamine restored learning in Wfs1∆Exon8 , Dizocilpine-treated, and Aβ25-35 -treated mice. These effects were observed at low ~1 mg/kg doses, not shared by Convolvine, the desmethyl metabolite, and blocked by an S1R antagonist. CONCLUSION AND IMPLICATIONS Convolamine therefore acts as an S1R positive modulator and this pharmacological action is relevant to the traditional use of Shankhpushpi in memory and cognitive protection.
Collapse
Affiliation(s)
- Lucie Crouzier
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France
| | - Johann Meunier
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France
| | - Allison Carles
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France
| | - Axelle Morilleau
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France
| | | | | | | | - Benjamin Delprat
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France
| | - Tangui Maurice
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France
| |
Collapse
|
4
|
Freyssin A, Carles A, Guehairia S, Rubinstenn G, Maurice T. Fluoroethylnormemantine (FENM) shows synergistic protection in combination with a sigma-1 receptor agonist in a mouse model of Alzheimer's disease. Neuropharmacology 2024; 242:109733. [PMID: 37844867 DOI: 10.1016/j.neuropharm.2023.109733] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2023] [Revised: 09/14/2023] [Accepted: 09/25/2023] [Indexed: 10/18/2023]
Abstract
Fluoroethylnormemantine (FENM) is a Memantine derivative with anti-amnesic and neuroprotective activities showed in the Aβ25-35 pharmacological mouse model of Alzheimer's disease (AD). As AD is a complex multi-factorial neurodegenerative pathology, combination therapies relying on drugs acting through different pathways, have been suggested to more adequately address neuroprotection. As several agonists of the sigma-1 receptor (S1R), an intracellular chaperone, are presently in phase 2 or 3 clinical trials in neurodegenetrative diseases including AD, we examined the potentialities of S1R drug-based combinations with FENM, or Memantine. Aβ25-35-treated mice were treated with S1R agonists (PRE-084, Igmesine, Cutamesine) and/or FENM, or Memantine, during 7 days after intracerebroventricular administration of oligomerized Aβ25-35. Mice were then tested for spatial short-term memory on day 8 and non-spatial long-term memory on days 9-10, using the spontaneous alternation or passive avoidance tests, respectively. The FENM or Memantine combination with Donepezil, that non-selectively inhibits acetylcholinesterase and activates S1R, was also tested. The efficacy of combinations using maximal non-active or minimal active doses of S1R agonist or FENM was analyzed using calculations of the combination index, based on simple isobologram representation. Data showed that most of the FENM-based combinations led to synergistic protection against Aβ25-35-induced learning deficits, for both long- and short-term memory responses, with a higher efficiency on the latter. Memantine led to synergistic combination in short-term memory but poorly in long-term memory responses, with either PRE-084 or Donepezil. These study showed that drug combinations based on FENM and S1R agonists may lead to highly effective and synergistic protection in AD, particularly on short-term memory.
Collapse
Affiliation(s)
- Aline Freyssin
- MMDN, Univ Montpellier, INSERM, EPHE, Montpellier, France; ReST Therapeutics, Montpellier, France
| | - Allison Carles
- MMDN, Univ Montpellier, INSERM, EPHE, Montpellier, France
| | | | | | - Tangui Maurice
- MMDN, Univ Montpellier, INSERM, EPHE, Montpellier, France.
| |
Collapse
|
5
|
Castle J, Shaw G, Weller D, Fielder E, Egnuni T, Singh M, Skinner R, von Zglinicki T, Clifford SC, Short SC, Miwa S, Hicks D. In vivo modeling recapitulates radiotherapy delivery and late-effect profile for childhood medulloblastoma. Neurooncol Adv 2024; 6:vdae091. [PMID: 38946880 PMCID: PMC11212071 DOI: 10.1093/noajnl/vdae091] [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] [Indexed: 07/02/2024] Open
Abstract
Background Medulloblastoma (MB) is the most common malignant pediatric brain tumor, with 5-year survival rates > 70%. Cranial radiotherapy (CRT) to the whole brain, with posterior fossa boost (PFB), underpins treatment for non-infants; however, radiotherapeutic insult to the normal brain has deleterious consequences to neurocognitive and physical functioning, and causes accelerated aging/frailty. Approaches to ameliorate radiotherapy-induced late-effects are lacking and a paucity of appropriate model systems hinders their development. Methods We have developed a clinically relevant in vivo model system that recapitulates the radiotherapy dose, targeting, and developmental stage of childhood medulloblastoma. Consistent with human regimens, age-equivalent (postnatal days 35-37) male C57Bl/6J mice received computerized tomography image-guided CRT (human-equivalent 37.5 Gy EQD2, n = 12) ± PFB (human-equivalent 48.7 Gy EQD2, n = 12), via the small animal radiation research platform and were longitudinally assessed for > 12 months. Results CRT was well tolerated, independent of PFB receipt. Compared to a sham-irradiated group (n = 12), irradiated mice were significantly frailer following irradiation (frailty index; P = .0002) and had reduced physical functioning; time to fall from a rotating rod (rotarod; P = .026) and grip strength (P = .006) were significantly lower. Neurocognitive deficits were consistent with childhood MB survivors; irradiated mice displayed significantly worse working memory (Y-maze; P = .009) and exhibited spatial memory deficits (Barnes maze; P = .029). Receipt of PFB did not induce a more severe late-effect profile. Conclusions Our in vivo model mirrored childhood MB radiotherapy and recapitulated features observed in the late-effect profile of MB survivors. Our clinically relevant model will facilitate both the elucidation of novel/target mechanisms underpinning MB late effects and the development of novel interventions for their amelioration.
Collapse
Affiliation(s)
- Jemma Castle
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Gary Shaw
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, Beckett St, Leeds, UK
| | - Dominic Weller
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Edward Fielder
- Biosciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Teklu Egnuni
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, Beckett St, Leeds, UK
| | - Mankaran Singh
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Roderick Skinner
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Thomas von Zglinicki
- Biosciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Steven C Clifford
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| | - Susan C Short
- Leeds Institute of Medical Research, Wellcome Trust Brenner Building, St James’s University Hospital, Beckett St, Leeds, UK
| | - Satomi Miwa
- Biosciences Institute, Campus for Ageing and Vitality, Newcastle University, Newcastle upon Tyne, UK
| | - Debbie Hicks
- Wolfson Childhood Cancer Research Centre, Newcastle University Centre for Cancer, Translational and Clinical Research Institute, Newcastle University, Newcastle upon Tyne, UK
| |
Collapse
|
6
|
Kim J, Kang H, Lee YB, Lee B, Lee D. A quantitative analysis of spontaneous alternation behaviors on a Y-maze reveals adverse effects of acute social isolation on spatial working memory. Sci Rep 2023; 13:14722. [PMID: 37679447 PMCID: PMC10485067 DOI: 10.1038/s41598-023-41996-4] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2023] [Accepted: 09/04/2023] [Indexed: 09/09/2023] Open
Abstract
Animals tend to alternate between different choices, which requires the ability to remember recent choices. The Y-maze spontaneous alternation test is widely used in various animal models for assessing short-term memory, and its precise evaluation depends upon the accurate determination of the arm visit sequence. However, an objective method for defining arm visits is lacking owing to uncertainty regarding the extent to which an animal must go into the arm to be considered visited. Here, we conducted quantitative analyses on mice behavior in the Y-maze while systematically varying the arm visit threshold and assessed the effect of acute social isolation on spatial working memory. Our results revealed that 24-h social isolation significantly reduced spontaneous alternation rate when the arm threshold was set at the distal part of the arm. Furthermore, the memory of the recently visited arms faded away faster in the socially isolated mice. However, other behavioral factors were comparable to those of the group-housed mice, indicating a specific impairment of short-term memory. Our findings suggest that the location of arm visit threshold is critical for the precise evaluation of short-term memory, and our study provides a method for comprehensively and systematically assessing spontaneous alternation behavior in the Y-maze.
Collapse
Affiliation(s)
- Joowon Kim
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Korea
- Graduate School of Medical Science and Engineering, Korea Advanced Institute of Science and Technology, Daejeon, 34141, Korea
| | - Hyeyeon Kang
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Korea
- Department of Biomedical Engineering, College of Information and Biotechnology, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Korea
| | - Young-Beom Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Korea
| | - Boyoung Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Korea
| | - Doyun Lee
- Center for Cognition and Sociality, Institute for Basic Science, Daejeon, 34126, Korea.
| |
Collapse
|
7
|
Godoy R, Macedo AB, Gervazio KY, Ribeiro LR, Lima JLF, Salvadori MGSS. Effects of ortho-eugenol on anxiety, working memory and oxidative stress in mice. BRAZ J BIOL 2023; 83:e271785. [PMID: 37610945 DOI: 10.1590/1519-6984.271785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2023] [Accepted: 06/21/2023] [Indexed: 08/25/2023] Open
Abstract
Ortho-eugenol is a synthetic derivative from eugenol, the major compound of clove essential oil, which has demonstrated antidepressant and antinociceptive effects in pioneering studies. Additionally, its effects appear to be dependent on the noradrenergic and dopaminergic systems. Depression and anxiety disorders are known to share a great overlap in their pathophysiology, and many drugs are effective in the treatment of both diseases. Furthermore, high levels of anxiety are related to working memory deficits and increased oxidative stress. Thus, in this study we investigated the effects of acute treatment of ortho-eugenol, at 50, 75 and 100 mg/kg, on anxiety, working memory and oxidative stress in male Swiss mice. Our results show that the 100 mg/kg dose increased the number of head-dips and reduced the latency in the hole-board test. The 50 mg/kg dose reduced malondialdehyde levels in the prefrontal cortex and the number of Y-maze entries compared to the MK-801-induced hyperlocomotion group. All doses reduced nitrite levels in the hippocampus. It was also possible to assess a statistical correlation between the reduction of oxidative stress and hyperlocomotion after the administration of ortho-eugenol. However, acute treatment was not able to prevent working memory deficits. Therefore, the present study shows that ortho-eugenol has an anxiolytic and antioxidant effect, and was able to prevent substance-induced hyperlocomotion. Our results contribute to the elucidation of the pharmacological profile of ortho-eugenol, as well as to direct further studies that seek to investigate its possible clinical applications.
Collapse
Affiliation(s)
- R Godoy
- Universidade Federal da Paraíba, Instituto de Pesquisa em Fármacos e Medicamentos, Laboratório de Psicofarmacologia, João Pessoa, PB, Brasil
| | - A B Macedo
- Universidade Federal da Paraíba, Instituto de Pesquisa em Fármacos e Medicamentos, Laboratório de Psicofarmacologia, João Pessoa, PB, Brasil
| | - K Y Gervazio
- Universidade Federal da Paraíba, Instituto de Pesquisa em Fármacos e Medicamentos, Laboratório de Psicofarmacologia, João Pessoa, PB, Brasil
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Programa de Pós-graduação em Produtos Bioativos Naturais e Sintéticos - PgPNSB, João Pessoa, PB, Brasil
| | - L R Ribeiro
- Universidade Federal da Paraíba, Instituto de Pesquisa em Fármacos e Medicamentos, Laboratório de Psicofarmacologia, João Pessoa, PB, Brasil
| | - J L F Lima
- Universidade Federal da Paraíba, Instituto de Pesquisa em Fármacos e Medicamentos, Laboratório de Psicofarmacologia, João Pessoa, PB, Brasil
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Programa de Pós-graduação em Produtos Bioativos Naturais e Sintéticos - PgPNSB, João Pessoa, PB, Brasil
| | - M G S S Salvadori
- Universidade Federal da Paraíba, Instituto de Pesquisa em Fármacos e Medicamentos, Laboratório de Psicofarmacologia, João Pessoa, PB, Brasil
- Universidade Federal da Paraíba, Centro de Ciências da Saúde, Programa de Pós-graduação em Produtos Bioativos Naturais e Sintéticos - PgPNSB, João Pessoa, PB, Brasil
| |
Collapse
|
8
|
Yen PSY, Liu YC, Chu CH, Chen SL. Upregulation of Glutamatergic Receptors in Hippocampus and Locomotor Hyperactivity in Aged Spontaneous Hypertensive Rat. Cell Mol Neurobiol 2022; 42:2205-2217. [PMID: 33954807 DOI: 10.1007/s10571-021-01094-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2020] [Accepted: 04/19/2021] [Indexed: 10/21/2022]
Abstract
Epidemiologic studies have indicated that chronic hypertension may facilitate the progression of abnormal behavior, such as emotional irritability, hyperactivity, and attention impairment. However, the mechanism of how chronic hypertension affects the brain and neuronal function remains unclear. In this study, 58-week-old male spontaneously hypertensive rats (SHR) and age-matched Wistar-Kyoto (WKY) control rats were used. Their locomotor activity and neuronal function were assessed by the open field test, novel object, and Y maze recognition test. Moreover brain tissues were analyzed. We found that the aged SHR exhibited significant locomotor hyperactivity when compared to the WKY rats. However, there was no significant difference in novel object and novel arm recognition between aged SHR and the WKY rats. In the analysis of synaptic membrane protein, the expression of glutamatergic receptors, such as the N-methyl-D-aspartate (NMDA) receptor receptors subunits 2B (GluN2B) and α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid (AMPA) receptor 1 (GluA1) in the hippocampus of SHR were significantly higher than those of WKY rats. In addition, in the synaptic membrane of SHR's hippocampus and medial prefrontal cortex (mPFC), a down-regulation of astrocytes was found, though the excitatory amino acid transporter 2 (EAAT2) remained constant. Moreover, a down-regulation of microglia in the hippocampus and mPFC was seen in the SHR brain. Long-term exposure to high blood pressure causes upregulation of glutamate receptors. The upregulation of glutamatergic receptors in hippocampus may contribute to the hyper-locomotor activity of aged rodents and may as a therapeutic target in hypertension-induced irritability and hyperactivity.
Collapse
Affiliation(s)
- Patrick Szu-Ying Yen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University (KMU), Kaohsiung, Taiwan
| | - Yen-Chin Liu
- Department of Anesthesiology, College of Medicine, National Cheng Kung University Hospital (NCKU), NCKU, 100 Shiquan 1st Rd, Sanmin Dist., Kaohsiung City, 807, Taiwan
| | - Chun-Hsien Chu
- Institute of Molecular Medicine, College of Medicine, NCKU, Tainan, Taiwan
| | - Shiou-Lan Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University (KMU), Kaohsiung, Taiwan.
- Department of Medical Research, College of Medicine, KMU Hospital & MSc Program in Tropical Medicine, KMU, Kaohsiung, Taiwan.
| |
Collapse
|
9
|
Carles A, Schlernitzauer A, Vignes M, Cros G, Magous R, Maurice T, Oiry C. Heptafluoroisobutyronitrile (C 4F 7N), a gas used for insulating and arc quenching in electrical switchgear, is neurotoxic in the mouse brain. Toxicology 2022; 480:153319. [PMID: 36100137 DOI: 10.1016/j.tox.2022.153319] [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: 07/22/2022] [Revised: 09/02/2022] [Accepted: 09/08/2022] [Indexed: 11/30/2022]
Abstract
Fluoronitrile gas (C4F7N, CAS number 42532-60-5) is one of the most promising candidates as insulating and/or breaking medium in high and medium voltage electrical equipment. Besides its promising properties, C4F7N gas is however not devoid of acute toxicity when used pure or in gas mixtures. The toxicity was not extensively analyzed and reported. The aim of the present study was to analyze in mice the consequences of a single exposure to C4F7N gas, at different concentrations and different timepoints after exposure. Male and female Swiss mice were exposed to breathable air or C4F7N gas, at 800 ppmv or 1500 ppmv, for 4 h on day 0. Behavioral tests (spontaneous alternation in the Y-maze and object recognition) were performed on days 1, 7 and 14 to assess memory alterations. The animals were then sacrificed and their brains dissected for biochemical analyses or fixed with paraformaldehyde for histology and immunohistochemistry. Results showed behavioral impairments and memory deficits, with impairments of alternation at days 1 and 7 and object recognition at day 14. Histological alterations of pyramidal neuronal layer in the hippocampus, neuroinflammatory astroglial reaction, and microglial alterations were observed, more marked in female than male mice. Moreover, the biochemical analyses done in the brain of 1500 ppmv exposed female mice showed a reductive stress with decreased lipid peroxidation and release of cytochrome c, leading to apoptosis with increases in caspase-9 cleavage and γ-H2AX/H2AX ratio. Finally, electrophysiological analyses using a multi-electrode array allowed the measure of the extracellular activity of pyramidal neurons in the CA2 area and revealed that exposure to the gas not only prevented the induction of long-term potentiation but even provoked an epileptoid-like activity in some neurons suggesting major alterations of synaptic plasticity. This study therefore showed that an acute exposure of mice to C4F7N gas provoked, particularly in female animals, memory alterations and brain toxicity characterized by a reductive stress, microglial toxicity, loss of synaptic plasticity and apoptosis. Its use in industrial installations must be done with extreme caution.
Collapse
Affiliation(s)
- Allison Carles
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France; ©MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France
| | | | - Michel Vignes
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Gérard Cros
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Richard Magous
- IBMM, Univ Montpellier, CNRS, ENSCM, Montpellier, France
| | - Tangui Maurice
- ©MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France.
| | - Catherine Oiry
- ©MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France.
| |
Collapse
|
10
|
Kim SR, Park Y, Li M, Kim YK, Lee S, Son SY, Lee S, Lee JS, Lee CH, Park HH, Lee JY, Hong S, Cho YC, Kim JW, Yoo HM, Cho N, Lee HS, Lee SH. Anti-inflammatory effect of Ailanthus altissima (Mill.) Swingle leaves in lipopolysaccharide-stimulated astrocytes. JOURNAL OF ETHNOPHARMACOLOGY 2022; 286:114258. [PMID: 34271112 DOI: 10.1016/j.jep.2021.114258] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/15/2021] [Revised: 05/18/2021] [Accepted: 05/25/2021] [Indexed: 06/13/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Activated astrocytes are involved in the progression of neurodegenerative diseases. Traditionally, Ailanthus altissima (Mill.) Swingle, widely distributed in East Asia, has been used as a medicine for the treatment of fever, gastric diseases, and inflammation. Although A. altissima has been reported to play an anti-inflammatory role in peripheral tissues or cells, its role in the central nervous system (CNS) remains unclear. AIM OF THE STUDY In the present study, we investigated the anti-inflammatory effects and mechanism of action of A. altissima in primary astrocytes stimulated by lipopolysaccharide (LPS). MATERIALS AND METHODS A nitrite assay was used to measure nitric oxide (NO) production, and the tetrazolium salt 3-(4, 5-dimethylthiazol-2-yl)-2, 5-diphenyltetrazolium bromide (MTT) assay was performed to determine cytotoxicity. The expression levels of inducible nitric oxide synthase (iNOS), cyclooxygenase-2 (COX-2), and mitogen-activated protein kinase (MAPK) were determined with western blotting. Reverse-transcription PCR was used to assess the expression of inflammatory cytokines. The levels of reactive oxygen species were measured using 2,7-dichlorodihydrofluorescein diacetate. Luciferase assay and immunocytochemistry were used for assessing nuclear factor-kappa B (NF-κB) transcription and p65 localization, respectively. Memory and social interaction were analyzed using the Y-maze and three-chamber tests, respectively. RESULTS The ethanol extract of A. altissima leaves (AAE) inhibited iNOS and COX-2 expression in LPS-stimulated astrocytes. Moreover, AAE reduced the transcription of various proinflammatory mediators, hindered NF-κB activation, and suppressed extracellular signal-regulated kinase (ERK) and c-Jun N-terminal kinase (JNK) activation without p38 activation. Ultra-high performance liquid chromatography with mass spectrometry analysis revealed that AAE comprised ethyl gallate, quercetin, and kaempferol, along with luteolin, which has anti-inflammatory properties, and repressed LPS-induced nitrite levels and the nuclear translocation of p65. Finally, oral administration of AAE attenuated LPS-induced memory and social impairment in mice and repressed LPS-induced ERK and JNK activation in the cortices of mice. CONCLUSION AAE could have therapeutic uses in the treatment of neuroinflammatory diseases via suppression of astrocyte activation.
Collapse
Affiliation(s)
- Sung Rae Kim
- Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Yongun Park
- Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Mo Li
- Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Yeong Kyeong Kim
- Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Sunmin Lee
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Su Young Son
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Sarah Lee
- National Institute of Biological Resources, Environmental Research Complex, 42 Hwangyeong-ro, Seo-gu, Incheon, 22755, Republic of Korea
| | - Jong Seok Lee
- National Institute of Biological Resources, Environmental Research Complex, 42 Hwangyeong-ro, Seo-gu, Incheon, 22755, Republic of Korea
| | - Choong Hwan Lee
- Department of Bioscience and Biotechnology, Konkuk University, 120 Neungdong-ro, Gwangjin-gu, Seoul, 05029, Republic of Korea
| | - Hyun Ho Park
- Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Ji-Yun Lee
- Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Sungguan Hong
- Department of Chemistry, Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea
| | - Young-Chang Cho
- College of Pharmacy, Chonnam National University, 77 Yongbong-ro, Buk-gu, Gwangju, 61186, Republic of Korea
| | - Jung-Woong Kim
- Department of Life Science, Chung-Ang University, Seoul, 06974, Republic of Korea
| | - Hee Min Yoo
- Center for Bioanalysis, Korea Research Institute of Standards and Science, Daejeon, 34113, Republic of Korea
| | - Namki Cho
- Research Institute of Pharmaceutical Sciences, College of Pharmacy, Chonnam National University, Gwangju, 61186, Republic of Korea
| | - Hyun-Shik Lee
- KNU-Center for Nonlinear Dynamics, CMRI, BK21 FOUR KNU Creative BioResearch Group, School of Life Sciences, College of Natural Sciences, Kyungpook National University, Daegu, 41566, Republic of Korea
| | - Sung Hoon Lee
- Chung-Ang University, 84 Heukseok-ro, Dongjak-gu, Seoul, 06974, Republic of Korea.
| |
Collapse
|
11
|
Martin P, Maurice T, Gammaitoni A, Farfel G, Boyd B, Galer B. Fenfluramine modulates the anti-amnesic effects induced by sigma-1 receptor agonists and neuro(active)steroids in vivo. Epilepsy Behav 2022; 127:108526. [PMID: 35007961 DOI: 10.1016/j.yebeh.2021.108526] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/19/2021] [Revised: 12/20/2021] [Accepted: 12/20/2021] [Indexed: 12/15/2022]
Abstract
Fenfluramine (N-ethyl-α-methl-3-(trifluoromethyl)phenethylamine) is an anti-seizure medication (ASM) particularly effective in patients with Dravet syndrome, a severe treatment-resistant epileptic encephalopathy. Fenfluramine acts not only as neuronal serotonin (5-HT) releaser but also as a positive modulator of the sigma-1 receptor (S1R). We here examined the modulatory activity of Fenfluramine on the S1R-mediated anti-amnesic response in mice using combination analyses. Fenfluramine and Norfenfluramine, racemate and isomers, were combined with either the S1R agonist (PRE-084) or the S1R-acting neuro(active)steroids, pregnenolone sulfate (PREGS), Dehydroepiandrosterone sulfate (DHEAS), or progesterone. We report that Fenfluramine racemate or (+)-Fenfluramine, in the 0.1-1 mg/kg dose range, attenuated the dizocilpine-induced learning deficits in spontaneous alternation and passive avoidance, and showed low-dose synergies in combination with PRE-084. These effects were blocked by the S1R antagonist NE-100. Dehydroepiandrosterone sulfate or PREGS attenuated dizocilpine-induced learning deficits in the 5-20 mg/kg dose range. Co-treatments at low dose between steroids and Fenfluramine or (+)-Fenfluramine were synergistic. Progesterone blocked Fenfluramine effect. Finally, Fenfluramine and (+)-Fenfluramine effects were prevented by the 5-HT1A receptor antagonist WAY-100635 or 5-HT2A antagonist RS-127445, but not by the 5-HT1B/1D antagonist GR 127935 or the 5-HT2C antagonist SB 242084, confirming a 5-HT1A and 5-HT2A receptor involvement in the drug effect on memory. We therefore confirmed the positive modulation of Fenfluramine racemate or dextroisomer on S1R and showed that, in physiological conditions, the drug potentiated the low dose effects of neuro(active)steroids, endogenous S1R modulators. The latter are potent modulators of the excitatory/inhibitory balance in the brain, and their levels must be considered in the antiepileptic action of Fenfluramine.
Collapse
Affiliation(s)
| | - Tangui Maurice
- MMDN, Univ Montpellier, EPHE, INSERM, Montpellier, France.
| | | | | | | | | |
Collapse
|
12
|
Abatematteo FS, Mosier PD, Niso M, Brunetti L, Berardi F, Loiodice F, Contino M, Delprat B, Maurice T, Laghezza A, Abate C. Development of novel phenoxyalkylpiperidines as high-affinity Sigma-1 (σ 1) receptor ligands with potent anti-amnesic effect. Eur J Med Chem 2022; 228:114038. [PMID: 34902734 DOI: 10.1016/j.ejmech.2021.114038] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 09/24/2021] [Accepted: 12/01/2021] [Indexed: 12/21/2022]
Abstract
The sigma-1 (σ1) receptor plays a significant role in many normal physiological functions and pathological disease states, and as such represents an attractive therapeutic target for both agonists and antagonists. Here, we describe a novel series of phenoxyalkylpiperidines based on the lead compound 1-[ω-(4-chlorophenoxy)ethyl]-4-methylpiperidine (1a) in which the degree of methylation at the carbon atoms alpha to the piperidine nitrogen was systematically varied. The affinity at σ1 and σ2 receptors and at Δ8-Δ7 sterol isomerase (SI) ranged from subnanomolar to micromolar Ki values. While the highest-affinity was displayed at the σ1, the increase of the degree of methylation in the piperidine ring progressively decreased the affinity. The subnanomolar affinity 1a and 1-[ω-(4-methoxyphenoxy)ethyl]-4-methylpiperidine (1b) displayed potent anti-amnesic effects associated with σ1 receptor agonism, in two memory tests. Automated receptor-small-molecule ligand docking provided a molecular structure-based rationale for the agonistic effects of 1a and 1b. Overall, the class of the phenoxyalkylpiperidines holds potential for the development of high affinity σ1 receptor agonists, and compound 1a, that appears as the best in class (exceeding by far the activity of the reference compound PRE-084) deserves further investigation.
Collapse
Affiliation(s)
- Francesca S Abatematteo
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari, ALDO MORO, Via Orabona, 4, I-70125, Bari, Italy
| | - Philip D Mosier
- Department of Biopharmaceutical Sciences, School of Pharmacy, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | - Mauro Niso
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari, ALDO MORO, Via Orabona, 4, I-70125, Bari, Italy
| | - Leonardo Brunetti
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari, ALDO MORO, Via Orabona, 4, I-70125, Bari, Italy
| | - Francesco Berardi
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari, ALDO MORO, Via Orabona, 4, I-70125, Bari, Italy
| | - Fulvio Loiodice
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari, ALDO MORO, Via Orabona, 4, I-70125, Bari, Italy
| | - Marialessandra Contino
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari, ALDO MORO, Via Orabona, 4, I-70125, Bari, Italy
| | - Benjamin Delprat
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France
| | - Tangui Maurice
- MMDN, University of Montpellier, EPHE, INSERM, Montpellier, France
| | - Antonio Laghezza
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari, ALDO MORO, Via Orabona, 4, I-70125, Bari, Italy.
| | - Carmen Abate
- Dipartimento di Farmacia-Scienze del Farmaco, Università degli Studi di Bari, ALDO MORO, Via Orabona, 4, I-70125, Bari, Italy.
| |
Collapse
|
13
|
Sałaciak K, Pytka K. Revisiting the sigma-1 receptor as a biological target to treat affective and cognitive disorders. Neurosci Biobehav Rev 2022; 132:1114-1136. [PMID: 34736882 PMCID: PMC8559442 DOI: 10.1016/j.neubiorev.2021.10.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/21/2022]
Abstract
Depression and cognitive disorders are diseases with complex and not-fully understood etiology. Unfortunately, the COVID-19 pandemic dramatically increased the prevalence of both conditions. Since the current treatments are inadequate in many patients, there is a constant need for discovering new compounds, which will be more effective in ameliorating depressive symptoms and treating cognitive decline. Proteins attracting much attention as potential targets for drugs treating these conditions are sigma-1 receptors. Sigma-1 receptors are multi-functional proteins localized in endoplasmic reticulum membranes, which play a crucial role in cellular signal transduction by interacting with receptors, ion channels, lipids, and kinases. Changes in their functions and expression may lead to various diseases, including depression or memory impairments. Thus, sigma-1 receptor modulation might be useful in treating these central nervous system diseases. Importantly, two sigma-1 receptor ligands entered clinical trials, showing that this compound group possesses therapeutic potential. Therefore, based on preclinical studies, this review discusses whether the sigma-1 receptor could be a promising target for drugs treating affective and cognitive disorders.
Collapse
Affiliation(s)
- Kinga Sałaciak
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland.
| |
Collapse
|
14
|
Martin P, Reeder T, Sourbron J, de Witte PAM, Gammaitoni AR, Galer BS. An Emerging Role for Sigma-1 Receptors in the Treatment of Developmental and Epileptic Encephalopathies. Int J Mol Sci 2021; 22:8416. [PMID: 34445144 PMCID: PMC8395113 DOI: 10.3390/ijms22168416] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2021] [Revised: 07/30/2021] [Accepted: 08/03/2021] [Indexed: 12/14/2022] Open
Abstract
Developmental and epileptic encephalopathies (DEEs) are complex conditions characterized primarily by seizures associated with neurodevelopmental and motor deficits. Recent evidence supports sigma-1 receptor modulation in both neuroprotection and antiseizure activity, suggesting that sigma-1 receptors may play a role in the pathogenesis of DEEs, and that targeting this receptor has the potential to positively impact both seizures and non-seizure outcomes in these disorders. Recent studies have demonstrated that the antiseizure medication fenfluramine, a serotonin-releasing drug that also acts as a positive modulator of sigma-1 receptors, reduces seizures and improves everyday executive functions (behavior, emotions, cognition) in patients with Dravet syndrome and Lennox-Gastaut syndrome. Here, we review the evidence for sigma-1 activity in reducing seizure frequency and promoting neuroprotection in the context of DEE pathophysiology and clinical presentation, using fenfluramine as a case example. Challenges and opportunities for future research include developing appropriate models for evaluating sigma-1 receptors in these syndromic epileptic conditions with multisystem involvement and complex clinical presentation.
Collapse
Affiliation(s)
- Parthena Martin
- Zogenix, Inc., Emeryville, CA 94608, USA; (P.M.); (T.R.); (A.R.G.)
| | - Thadd Reeder
- Zogenix, Inc., Emeryville, CA 94608, USA; (P.M.); (T.R.); (A.R.G.)
| | - Jo Sourbron
- University Hospital KU Leuven, 3000 Leuven, Belgium;
| | - Peter A. M. de Witte
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences at KU Leuven, 3000 Leuven, Belgium;
| | | | - Bradley S. Galer
- Zogenix, Inc., Emeryville, CA 94608, USA; (P.M.); (T.R.); (A.R.G.)
| |
Collapse
|
15
|
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: 34] [Impact Index Per Article: 11.3] [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
|
16
|
Chiu YJ, Lin CH, Lee MC, Hsieh-Li HM, Chen CM, Wu YR, Chang KH, Lee-Chen GJ. Formulated Chinese medicine Shaoyao Gancao Tang reduces NLRP1 and NLRP3 in Alzheimer's disease cell and mouse models for neuroprotection and cognitive improvement. Aging (Albany NY) 2021; 13:15620-15637. [PMID: 34106880 PMCID: PMC8221334 DOI: 10.18632/aging.203125] [Citation(s) in RCA: 32] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2020] [Accepted: 03/23/2021] [Indexed: 12/18/2022]
Abstract
Amyloid β (Aβ) plays a major role in the neurodegeneration of Alzheimer’s disease (AD). The accumulation of misfolded Aβ causes oxidative stress and inflammatory damage leading to apoptotic cell death. Traditional Chinese herbal medicine (CHM) has been widely used in treating neurodegenerative diseases by reducing oxidative stress and neuroinflammation. We examined the neuroprotective effect of formulated CHM Shaoyao Gancao Tang (SG-Tang, made of Paeonia lactiflora and Glycyrrhiza uralensis at 1:1 ratio) in AD cell and mouse models. In Aβ-GFP SH-SY5Y cells, SG-Tang reduced Aβ aggregation and reactive oxygen species (ROS) production, as well as improved neurite outgrowth. When the Aβ-GFP-expressing cells were stimulated with conditioned medium from interferon (IFN)-γ-activated HMC3 microglia, SG-Tang suppressed expressions of inducible nitric oxide synthase (iNOS), NLR family pyrin domain containing 1 (NLRP1) and 3 (NLRP3), tumor necrosis factor (TNF)-α, interleukin (IL)-1β and IL-6, attenuated caspase-1 activity and ROS production, and promoted neurite outgrowth. In streptozocin-induced hyperglycemic APP/PS1/Tau triple transgenic (3×Tg-AD) mice, SG-Tang also reduced expressions of NLRP1, NLRP3, Aβ and Tau in hippocampus and cortex, as well as improved working and spatial memories in Y maze and Morris water maze. Collectively, our results demonstrate the potential of SG-Tang in treating AD by moderating neuroinflammation.
Collapse
Affiliation(s)
- Ya-Jen Chiu
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Chih-Hsin Lin
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Ming-Chung Lee
- Sun Ten Pharmaceutical Co. Ltd., New Taipei City 23143, Taiwan
| | - Hsiu Mei Hsieh-Li
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Kuo-Hsuan Chang
- Department of Neurology, Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Taoyuan 33302, Taiwan
| | - Guey-Jen Lee-Chen
- Department of Life Science, National Taiwan Normal University, Taipei 11677, Taiwan
| |
Collapse
|
17
|
Takamura Y, Kakuta H. In Vivo Receptor Visualization and Evaluation of Receptor Occupancy with Positron Emission Tomography. J Med Chem 2021; 64:5226-5251. [PMID: 33905258 DOI: 10.1021/acs.jmedchem.0c01714] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Positron emission tomography (PET) is useful for noninvasive in vivo visualization of disease-related receptors, for evaluation of receptor occupancy to determine an appropriate drug dosage, and for proof-of-concept of drug candidates in translational research. For these purposes, the specificity of the PET tracer for the target receptor is critical. Here, we review work in this area, focusing on the chemical structures of reported PET tracers, their Ki/Kd values, and the physical properties relevant to target receptor selectivity. Among these physical properties, such as cLogP, cLogD, molecular weight, topological polar surface area, number of hydrogen bond donors, and pKa, we focus especially on LogD and LogP as important physical properties that can be easily compared across a range of studies. We discuss the success of PET tracers in evaluating receptor occupancy and consider likely future developments in the field.
Collapse
Affiliation(s)
- Yuta Takamura
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| | - Hiroki Kakuta
- Division of Pharmaceutical Sciences, Okayama University Graduate School of Medicine, Dentistry and Pharmaceutical Sciences, 1-1-1, Tsushima-naka, Kita-ku, Okayama 700-8530, Japan
| |
Collapse
|
18
|
Maurice T. Bi-phasic dose response in the preclinical and clinical developments of sigma-1 receptor ligands for the treatment of neurodegenerative disorders. Expert Opin Drug Discov 2021; 16:373-389. [PMID: 33070647 DOI: 10.1080/17460441.2021.1838483] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Accepted: 10/14/2020] [Indexed: 12/19/2022]
Abstract
Introduction: The sigma-1 receptor (S1R) is attracting much attention for disease-modifying therapies in neurodegenerative diseases. It is a conserved protein, present in plasma and endoplasmic reticulum (ER) membranes and enriched in mitochondria-associated ER membranes (MAMs). It modulates ER-mitochondria Ca2+ transfer and ER stress pathways. Mitochondrial and MAM dysfunctions contribute to neurodegenerative processes in diseases such as Alzheimer, Parkinson, Huntington or Amyotrophic Lateral Sclerosis. Interestingly, the S1R can be activated by small druggable molecules and accumulating preclinical data suggest that S1R agonists are effective protectants in these neurodegenerative diseases.Area covered: In this review, we will present the data showing the high therapeutic potential of S1R drugs for the treatment of neurodegenerative diseases, focusing on pridopidine as a potent and selective S1R agonist under clinical development. Of particular interest is the bi-phasic (bell-shaped) dose-response effect, representing a common feature of all S1R agonists and described in numerous preclinical models in vitro, in vivo and in clinical trials.Expert opinion: S1R agonists modulate inter-organelles communication altered in neurodegenerative diseases and activate intracellular survival pathways. Research will continue growing in the future. The particular cellular nature of this chaperone protein must be better understood to facilitate the clinical developement of promising molecules.
Collapse
Affiliation(s)
- Tangui Maurice
- MMDN, Univ Montpellier, EPHE, INSERM, UMR_S1198, Montpellier, France
| |
Collapse
|
19
|
Lin YS, Liu CK, Lee HC, Chou MC, Ke LY, Chen CH, Chen SL. Electronegative very-low-density lipoprotein induces brain inflammation and cognitive dysfunction in mice. Sci Rep 2021; 11:6013. [PMID: 33727609 PMCID: PMC7966811 DOI: 10.1038/s41598-021-85502-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/06/2020] [Accepted: 03/02/2021] [Indexed: 11/09/2022] Open
Abstract
Epidemiologic studies have indicated that dyslipidemia may facilitate the progression of cognitive dysfunction. We previously showed that patients with metabolic syndrome (MetS) had significantly higher plasma levels of electronegative very-low-density lipoprotein (VLDL) than did healthy controls. However, the effects of electronegative-VLDL on the brain and cognitive function remain unclear. In this study, VLDL isolated from healthy volunteers (nVLDL) or patients with MetS (metVLDL) was administered to mice by means of tail vein injection. Cognitive function was assessed by using the Y maze test, and plasma and brain tissues were analyzed. We found that mice injected with metVLDL but not nVLDL exhibited significant hippocampus CA3 neuronal cell loss and cognitive dysfunction. In mice injected with nVLDL, we observed mild glial cell activation in the medial prefrontal cortex (mPFC) and hippocampus CA3. However, in mice injected with metVLDL, plasma and brain TNF-α and Aβ-42 levels and glial cell activation in the mPFC and whole hippocampus were higher than those in control mice. In conclusion, long-term exposure to metVLDL induced levels of TNF-α, Aβ-42, and glial cells in the brain, contributing to the progression of cognitive dysfunction. Our findings suggest that electronegative-VLDL levels may represent a new therapeutic target for cognitive dysfunction.
Collapse
Affiliation(s)
- Ying-Shao Lin
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University (KMU), 100 Shiquan 1st Rd, Sanmin Dist., Kaohsiung City, 807, Taiwan
| | - Ching-Kuan Liu
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University (KMU), 100 Shiquan 1st Rd, Sanmin Dist., Kaohsiung City, 807, Taiwan.,Department of Neurology, KMU Hospital, KMU, Kaohsiung, Taiwan.,Department of Neurology, Faculty of Medicine, College of Medicine, KMU, Kaohsiung, Taiwan
| | - Hsiang-Chun Lee
- Division of Cardiology, Department of Internal Medicine, KMU Hospital and Faculty of Medicine, College of Medicine, KMU, Kaohsiung, Taiwan.,Lipid Science and Aging Research Center, College of Medicine, KMU, Kaohsiung, Taiwan
| | - Mei-Chuan Chou
- Department of Neurology, KMU Hospital, KMU, Kaohsiung, Taiwan.,Graduate Institute of Clinical Medicine, College of Medicine, KMU, Kaohsiung, Taiwan.,Department of Neurology, Kaohsiung Municipal Ta-Tung Hospital, KMU, Kaohsiung, Taiwan
| | - Liang-Yin Ke
- Lipid Science and Aging Research Center, College of Medicine, KMU, Kaohsiung, Taiwan.,Department of Medical Laboratory Science and Biotechnology, KMU, Kaohsiung, Taiwan
| | - Chu-Huang Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University (KMU), 100 Shiquan 1st Rd, Sanmin Dist., Kaohsiung City, 807, Taiwan.,Lipid Science and Aging Research Center, College of Medicine, KMU, Kaohsiung, Taiwan.,Vascular and Medicinal Research, Texas Heart Institute, Houston, TX, USA
| | - Shiou-Lan Chen
- Graduate Institute of Medicine, College of Medicine, Kaohsiung Medical University (KMU), 100 Shiquan 1st Rd, Sanmin Dist., Kaohsiung City, 807, Taiwan. .,Department of Medical Research, KMU Hospital, Drug Development and Value Creation Research Center and MSc Program in Tropical Medicine, KMU, Kaohsiung, Taiwan.
| |
Collapse
|
20
|
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: 1.0] [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
|
21
|
Limón ID, Angulo-Cruz I, Sánchez-Abdon L, Patricio-Martínez A. Disturbance of the Glutamate-Glutamine Cycle, Secondary to Hepatic Damage, Compromises Memory Function. Front Neurosci 2021; 15:578922. [PMID: 33584185 PMCID: PMC7873464 DOI: 10.3389/fnins.2021.578922] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2020] [Accepted: 01/06/2021] [Indexed: 12/13/2022] Open
Abstract
Glutamate fulfils many vital functions both at a peripheral level and in the central nervous system (CNS). However, hyperammonemia and hepatic failure induce alterations in glutamatergic neurotransmission, which may be the main cause of hepatic encephalopathy (HE), an imbalance which may explain damage to both learning and memory. Cognitive and motor alterations in hyperammonemia may be caused by a deregulation of the glutamate-glutamine cycle, particularly in astrocytes, due to the blocking of the glutamate excitatory amino-acid transporters 1 and 2 (EAAT1, EAAT2). Excess extracellular glutamate triggers mechanisms involving astrocyte-mediated inflammation, including the release of Ca2+-dependent glutamate from astrocytes, the appearance of excitotoxicity, the formation of reactive oxygen species (ROS), and cell damage. Glutamate re-uptake not only prevents excitotoxicity, but also acts as a vital component in synaptic plasticity and function. The present review outlines the evidence of the relationship between hepatic damage, such as that occurring in HE and hyperammonemia, and changes in glutamine synthetase function, which increase glutamate concentrations in the CNS. These conditions produce dysfunction in neuronal communication. The present review also includes data indicating that hyperammonemia is related to the release of a high level of pro-inflammatory factors, such as interleukin-6, by astrocytes. This neuroinflammatory condition alters the function of the membrane receptors, such as N-methyl-D-aspartate (NMDA), (α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid) AMPA, and γ-aminobutyric acid (GABA), thus affecting learning and spatial memory. Data indicates that learning and spatial memory, as well as discriminatory or other information acquisition processes in the CNS, are damaged by the appearance of hyperammonemia and, moreover, are associated with a reduction in the production of cyclic guanosine monophosphate (cGMP). Therefore, increased levels of pharmacologically controlled cGMP may be used as a therapeutic tool for improving learning and memory in patients with HE, hyperammonemia, cerebral oedema, or reduced intellectual capacity.
Collapse
Affiliation(s)
| | - Isael Angulo-Cruz
- Laboratorio de Neurofarmacología, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Lesli Sánchez-Abdon
- Laboratorio de Neurofarmacología, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| | - Aleidy Patricio-Martínez
- Laboratorio de Neurofarmacología, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
- Facultad de Ciencias Biológicas, Benemérita Universidad Autónoma de Puebla, Puebla, Mexico
| |
Collapse
|
22
|
Shin T, Hiraoka Y, Yamasaki T, Marth JD, Penninger JM, Kanai-Azuma M, Tanaka K, Kofuji S, Nishina H. MKK7 deficiency in mature neurons impairs parental behavior in mice. Genes Cells 2020; 26:5-17. [PMID: 33098150 PMCID: PMC7839552 DOI: 10.1111/gtc.12816] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/08/2020] [Revised: 10/15/2020] [Accepted: 10/16/2020] [Indexed: 11/28/2022]
Abstract
c‐Jun N‐terminal kinases (JNKs) are constitutively activated in mammalian brains and are indispensable for their development and neural functions. MKK7 is an upstream activator of all JNKs. However, whether the common JNK signaling pathway regulates the brain's control of social behavior remains unclear. Here, we show that female mice in which Mkk7 is deleted specifically in mature neurons (Mkk7flox/floxSyn‐Cre mice) give birth to a normal number of pups but fail to raise them due to a defect in pup retrieval. To explore the mechanism underlying this abnormality, we performed comprehensive behavioral tests. Mkk7flox/floxSyn‐Cre mice showed normal locomotor functions and cognitive ability but exhibited depression‐like behavior. cDNA microarray analysis of mutant brain revealed an altered gene expression pattern. Quantitative RT‐PCR analysis demonstrated that mRNA expression levels of genes related to neural signaling pathways and a calcium channel were significantly different from controls. In addition, loss of neural MKK7 had unexpected regulatory effects on gene expression patterns in oligodendrocytes. These findings indicate that MKK7 has an important role in regulating the gene expression patterns responsible for promoting normal social behavior and staving off depression.
Collapse
Affiliation(s)
- Tadashi Shin
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Yuichi Hiraoka
- Department of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Tokiwa Yamasaki
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan.,Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Jamey D Marth
- Center for Nanomedicine, Department of Molecular, Cellular and Developmental Biology, SBP Medical Discovery Institute, University of California Santa Barbara, Santa Barbara, CA, USA
| | - Josef M Penninger
- IMBA, Institute of Molecular Biotechnology of the Austrian Academy of Sciences, Vienna, Austria.,Department of Medical Genetics, Life Sciences Institute, University of British Columbia, Vancouver, BC, Canada
| | - Masami Kanai-Azuma
- Department of Experimental Animal Model for Human Disease, Center for Experimental Animals, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Kohichi Tanaka
- Department of Molecular Neuroscience, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Satoshi Kofuji
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| | - Hiroshi Nishina
- Department of Developmental and Regenerative Biology, Medical Research Institute, Tokyo Medical and Dental University (TMDU), Tokyo, Japan
| |
Collapse
|
23
|
Gudapati K, Singh A, Clarkson-Townsend D, Feola AJ, Allen RS. Behavioral Assessment of Visual Function via Optomotor Response and Cognitive Function via Y-Maze in Diabetic Rats. J Vis Exp 2020. [PMID: 33165321 DOI: 10.3791/61806] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
The optomotor response and the Y-maze are behavioral tests useful for assessing visual and cognitive function, respectively. The optomotor response is a valuable tool to track changes in spatial frequency (SF) and contrast sensitivity (CS) thresholds over time in a number of retinal disease models, including diabetic retinopathy. Similarly, the Y-maze can be used to monitor spatial cognition (as measured by spontaneous alternation) and exploratory behavior (as measured by a number of entries) in a number of disease models that affect the central nervous system. Advantages of the optomotor response and the Y-maze include sensitivity, speed of testing, the use of innate responses (training is not needed), and the ability to be performed on awake (non-anesthetized) animals. Here, protocols are described for both the optomotor response and the Y-maze and examples of their use shown in models of Type I and Type II diabetes. Methods include preparation of rodents and equipment, performance of the optomotor response and the Y-maze, and post-test data analysis.
Collapse
Affiliation(s)
- Kaavya Gudapati
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center; Department of Biomedical Engineering, Georgia Institute of Technology
| | - Anayesha Singh
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center; Department of Neuroscience, Emory University
| | - Danielle Clarkson-Townsend
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center; Gangarosa Department of Environmental Health, Emory University
| | - Andrew J Feola
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center; Department of Biomedical Engineering, Georgia Institute of Technology
| | - Rachael S Allen
- Center for Visual and Neurocognitive Rehabilitation, Atlanta VA Medical Center; Department of Biomedical Engineering, Georgia Institute of Technology;
| |
Collapse
|
24
|
Brimson JM, Brimson S, Chomchoei C, Tencomnao T. Using sigma-ligands as part of a multi-receptor approach to target diseases of the brain. Expert Opin Ther Targets 2020; 24:1009-1028. [PMID: 32746649 DOI: 10.1080/14728222.2020.1805435] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The sigma receptors are found abundantly in the central nervous system and are targets for the treatment of various diseases, including Alzheimer's (AD), Parkinson's (PD), Huntington's disease (HD), depression, amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). However, for many of these diseases, other receptors and targets have been the focus of the most, such as acetylcholine esterase inhibitors in Alzheimer's and dopamine replacement in Parkinson's. The currently available drugs for these diseases have limited success resulting in the requirement of an alternative approach to their treatment. AREAS COVERED In this review, we discuss the potential role of the sigma receptors and their ligands as part of a multi receptor approach in the treatment of the diseases mentioned above. The literature reviewed was obtained through searches in databases, including PubMed, Web of Science, Google Scholar, and Scopus. EXPERT OPINION Given sigma receptor agonists provide neuroprotection along with other benefits such as potentiating the effects of other receptors, further development of multi-receptor targeting ligands, and or the development of multi-drug combinations to target multiple receptors may prove beneficial in the future treatment of degenerative diseases of the CNS, especially when coupled with better diagnostic techniques.
Collapse
Affiliation(s)
- James Michael Brimson
- Age-related Inflammation and Degeneration Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University , Bangkok, Thailand
| | - Sirikalaya Brimson
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University , Bangkok, Thailand
| | - Chanichon Chomchoei
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University , Bangkok, Thailand
| | - Tewin Tencomnao
- Age-related Inflammation and Degeneration Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University , Bangkok, Thailand
| |
Collapse
|
25
|
Rolfes S, Munro DAD, Lyras EM, Matute E, Ouk K, Harms C, Böttcher C, Priller J. Lentiviral delivery of human erythropoietin attenuates hippocampal atrophy and improves cognition in the R6/2 mouse model of Huntington's disease. Neurobiol Dis 2020; 144:105024. [PMID: 32702387 DOI: 10.1016/j.nbd.2020.105024] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Revised: 07/04/2020] [Accepted: 07/16/2020] [Indexed: 12/13/2022] Open
Abstract
Huntington's disease (HD) is an incurable neurodegenerative disorder caused by a trinucleotide (CAG) repeat expansion in the huntingtin gene (HTT). The R6/2 transgenic mouse model of HD expresses exon 1 of the human HTT gene with approximately 150 CAG repeats. R6/2 mice develop progressive behavioural abnormalities, impaired neurogenesis, and atrophy of several brain regions. In recent years, erythropoietin (EPO) has been shown to confer neuroprotection and enhance neurogenesis, rendering it a promising molecule to attenuate HD symptoms. In this study, the therapeutic potential of EPO was evaluated in female R6/2 transgenic mice. A single bilateral injection of a lentivirus encoding human EPO (LV-hEPO) was performed into the lateral ventricles of R6/2 mice at disease onset (8 weeks of age). Control groups were either untreated or injected with a lentivirus encoding green fluorescent protein (LV-GFP). Thirty days after virus administration, hEPO mRNA and protein were present in injected R6/2 brains. Compared to control R6/2 mice, LV-hEPO-treated R6/2 mice exhibited reduced hippocampal atrophy, increased neuroblast branching towards the dentate granular cell layer, and improved spatial cognition. Our results suggest that LV-hEPO administration may be a promising strategy to reduce cognitive impairment in HD.
Collapse
Affiliation(s)
- Simone Rolfes
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité -Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - David A D Munro
- UK Dementia Research Institute at the University of Edinburgh, Edinburgh EH16 4SA, UK
| | - Ekaterini-Maria Lyras
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité -Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Eduardo Matute
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité -Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Koliane Ouk
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité -Universitätsmedizin Berlin, 10117 Berlin, Germany; DZNE Berlin, 10117 Berlin, Germany
| | - Christoph Harms
- Department of Experimental Neurology and Center for Stroke Research, Charité -Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Chotima Böttcher
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité -Universitätsmedizin Berlin, 10117 Berlin, Germany
| | - Josef Priller
- Department of Neuropsychiatry and Laboratory of Molecular Psychiatry, Charité -Universitätsmedizin Berlin, 10117 Berlin, Germany; UK Dementia Research Institute at the University of Edinburgh, Edinburgh EH16 4SA, UK; DZNE Berlin, 10117 Berlin, Germany.
| |
Collapse
|
26
|
Rodrigues KDS, Klein CP, August PM, Dos Santos BG, Hözer RM, Maurmann RM, Scortegagna MC, Hoppe JB, Matté C. Early weaning alters redox status in the hippocampus and hypothalamus of rat pups. Int J Dev Neurosci 2020; 80:512-527. [PMID: 32619317 DOI: 10.1002/jdn.10047] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2020] [Revised: 06/15/2020] [Accepted: 06/23/2020] [Indexed: 12/17/2022] Open
Abstract
Exposure to environmental factors can program the metabolism, conferring resistance or increasing the risk to chronic disease development in childhood and adulthood. In this sense, lactation is an important period in this window of development. Herein, we investigated the effect of early weaning on neurochemical and behavioral changes in offspring at weaning and adulthood. Female and male pups were divided into four groups: (1) Control weaning (weaning on the PND21, pups were kept with the biological mother); (2) Early Weaning Bromocriptine group (EWB) (pharmacological weaning on PND16); (3) Early Weaning Cross-Fostering group (EWCF) (pups housed with a foster mother on PND16 up to PND21); (4) Early Weaning Without Care group (EWWC) (weaning on PND16, maternal separation). Weight control of pups was recorded from postnatal Day 16 to 59. On the 21st day, part of the pups was euthanized and the hippocampus and hypothalamus were removed for biochemical evaluation. The remaining pups were submitted to behavioral tests on the 60th postnatal day. Early weaning reduced the pups' body weight, in a sex-dependent way. At 60 days of age, male pups of EWCF and EWWC groups have lower body weight compared to control male, and female body weight was lower than male pups. In relation to biochemical changes in the brain, weaning altered the levels of oxidants, increased the enzymatic activity of superoxide dismutase (SOD), and glutathione peroxidase (GPx), as well as induced lipid peroxidation. Weaning was also able to alter long-term memory and induce anxious behavior in pups. Our results demonstrate that the different types of early weaning changed the parameters of redox status in the hippocampus and hypothalamus of pups (21 days old), suggesting a prooxidative profile, in addition, to alter learning/memory and inducing an anxious behavior in male offspring (60 days old).
Collapse
Affiliation(s)
- Karoline Dos Santos Rodrigues
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Caroline Peres Klein
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Pauline Maciel August
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Bernardo Gindri Dos Santos
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Régis Mateus Hözer
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Rafael Moura Maurmann
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Mariana Crestani Scortegagna
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Juliana Bender Hoppe
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| | - Cristiane Matté
- Programa de Pós-graduação em Ciências Biológicas: Bioquímica, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil.,Programa de Pós-graduação em Ciências Biológicas: Fisiologia, ICBS, Universidade Federal do Rio Grande do Sul, Porto Alegre, Brazil
| |
Collapse
|
27
|
Martin P, de Witte PAM, Maurice T, Gammaitoni A, Farfel G, Galer B. Fenfluramine acts as a positive modulator of sigma-1 receptors. Epilepsy Behav 2020; 105:106989. [PMID: 32169824 DOI: 10.1016/j.yebeh.2020.106989] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/11/2019] [Revised: 02/10/2020] [Accepted: 02/17/2020] [Indexed: 12/14/2022]
Abstract
OBJECTIVE Adjunctive fenfluramine hydrochloride, classically described as acting pharmacologically through a serotonergic mechanism, has demonstrated a unique and robust clinical response profile with regard to its magnitude, consistency, and durability of effect on seizure activity in patients with pharmacoresistant Dravet syndrome. Recent findings also support long-term improvements in executive functions (behavior, emotion, cognition) in these patients. The observed clinical profile is inconsistent with serotonergic activity alone, as other serotonergic medications have not been demonstrated to have these clinical effects. This study investigated a potential role for σ1 receptor activity in complementing fenfluramine's serotonergic pharmacology. METHODS Radioligand binding assays tested the affinity of fenfluramine for 47 receptors associated with seizures in the literature, including σ receptors. Cellular function assays tested fenfluramine and norfenfluramine (its major metabolite) activity at various receptors, including adrenergic, muscarinic, and serotonergic receptors. The σ1 receptor activity was assessed by the mouse vas deferens isometric twitch and by an assay of dissociation of the σ1 receptor from the endoplasmic reticulum stress protein binding immunoglobulin protein (BiP). In vivo mouse models assessed fenfluramine activity at σ1 receptors in ameliorating dizocilpine-induced learning deficits in spatial and nonspatial memory tasks, alone or in combination with the reference σ1 receptor agonist PRE-084. RESULTS Fenfluramine and norfenfluramine bound ≥30% to β2-adrenergic, muscarinic M1, serotonergic 5-HT1A, and σ receptors, as well as sodium channels, with a Ki between 266 nM (σ receptors) and 17.5 μM (β-adrenergic receptors). However, only σ1 receptor isometric twitch assays showed a positive functional response, with weak stimulation by fenfluramine and inhibition by norfenfluramine. Fenfluramine, but not the 5-HT2C agonist lorcaserin, showed a positive modulation of the PRE-084-induced dissociation of σ1 protein from BiP. Fenfluramine also showed dose-dependent antiamnesic effects against dizocilpine-induced learning deficits in spontaneous alternation and passive avoidance responses, which are models of σ1 activation. Moreover, low doses of fenfluramine synergistically potentiated the low-dose effect of PRE-084, confirming a positive modulatory effect at the σ1 receptor. Finally, all in vivo effects were blocked by the σ1 receptor antagonist NE-100. SIGNIFICANCE Fenfluramine demonstrated modulatory activity at σ1 receptors in vitro and in vivo in addition to its known serotonergic activity. These studies identify a possible new σ1 receptor mechanism underpinning fenfluramine's central nervous system effects, which may contribute to its antiseizure activity in Dravet syndrome and positive effects observed on executive functions in clinical studies.
Collapse
Affiliation(s)
| | - Peter A M de Witte
- Laboratory for Molecular Biodiscovery, Department of Pharmaceutical and Pharmacological Sciences, KU Leuven, Leuven, Belgium.
| | - Tangui Maurice
- MMDN, University of Montpellier, INSERM, EPHE, UMR_S1198, Montpellier, France.
| | | | | | | |
Collapse
|
28
|
McGarry A, Leinonen M, Kieburtz K, Geva M, Olanow CW, Hayden M. Effects of Pridopidine on Functional Capacity in Early-Stage Participants from the PRIDE-HD Study. J Huntingtons Dis 2020; 9:371-380. [PMID: 33164941 PMCID: PMC7836066 DOI: 10.3233/jhd-200440] [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] [Indexed: 02/03/2023]
Abstract
BACKGROUND No pharmacological treatment has been demonstrated to provide a functional benefit for persons with Huntington's disease (HD). Pridopidine is a sigma-1-receptor agonist shown to have beneficial effects in preclinical models of HD. OBJECTIVE To further explore the effect of pridopidine on Total Functional Capacity (TFC) in the recent double-blind, placebo-controlled PRIDE-HD study. METHODS We performed post-hoc analyses to evaluate the effect of pridopidine on TFC at 26 and 52 weeks. Participants were stratified according to baseline TFC score and analyzed using repeated measures (MMRM) and multiple imputation assuming missing not-at-random (MNAR) and worst-case scenarios. RESULTS The pridopidine 45 mg bid dosage demonstrated a beneficial effect on TFC for the entire population at week 52 of 0.87 (nominal p = 0.0032). The effect was more pronounced for early HD participants (HD1/HD2, TFC = 7-13), with a change from placebo of 1.16 (nominal p = 0.0003). This effect remained nominally significant using multiple imputation with missing not at random assumption as a sensitivity analysis. Responder analyses showed pridopidine 45 mg bid reduced the probability of TFC decline in early HD patients at Week 52 (nominal p = 0.02). CONCLUSION Pridopidine 45 mg bid results in a nominally significant reduction in TFC decline at 52 weeks compared to placebo, particularly in patients with early-stage HD.
Collapse
Affiliation(s)
- Andrew McGarry
- Cooper University Healthcare at Rowan University, Camden, NJ, USA
- Clintrex Research Corporation, Sarasota, FL, USA
| | | | | | | | - C. Warren Olanow
- Mount Sinai School of Medicine, New York City, NY, USA
- Clintrex Research Corporation, Sarasota, FL, USA
| | - Michael Hayden
- Prilenia Therapeutics, Herzliya, Israel
- Centre for Molecular Medicine and Therapeutics, University of British Columbia, Vancouver, BC, Canada
| |
Collapse
|
29
|
Todorovic ST, Smiljanic KR, Ruzdijic SD, Djordjevic ANM, Kanazir SD. Effects of Different Dietary Protocols on General Activity and Frailty of Male Wistar Rats During Aging. J Gerontol A Biol Sci Med Sci 2019; 73:1036-1044. [PMID: 29415252 DOI: 10.1093/gerona/gly015] [Citation(s) in RCA: 25] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2017] [Accepted: 01/31/2018] [Indexed: 01/22/2023] Open
Abstract
Dietary restriction (DR) is an important experimental paradigm for lifespan and healthspan extension, but its specific contribution regarding the type, onset, and duration are still debatable. This study was designed to examine the impact of different dietary protocols by assessing the behavioral changes during aging. We exposed male Wistar rats of various age to ad libitum (AL) or DR (60 per cent of AL daily intake) feeding regimens with different onsets. The impact of DR on locomotor activity, memory, and learning was examined in 12-, 18-, and 24-month-old treated animals and controls using open field and Y-maze tests. We have also evaluated the effects of different DR's through the quantification of animal frailty, using behavioral data to create the frailty score. Our results indicated that DR improves general animal activity and spatial memory and decreases frailty with the effect being highly dependent on DR duration and onset. Notably, life-long restriction started at young age had the most profound effect. In contrast, shorter duration and later onset of restricted diet had significantly lower or no impact on animal's behavior and frailty. This study signifies the importance of DR starting point and duration as critical determinants of DR effects on healthspan.
Collapse
Affiliation(s)
- Smilja T Todorovic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic," University of Belgrade, Belgrade, Serbia
| | - Kosara R Smiljanic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic," University of Belgrade, Belgrade, Serbia
| | - Sabera D Ruzdijic
- Department of Neurobiology, Institute for Biological Research "Sinisa Stankovic," University of Belgrade, Belgrade, Serbia
| | | | | |
Collapse
|
30
|
Knockdown of the CXCL12/CXCR7 chemokine pathway results in learning deficits and neural progenitor maturation impairment in mice. Brain Behav Immun 2019; 80:697-710. [PMID: 31100368 DOI: 10.1016/j.bbi.2019.05.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 05/10/2019] [Accepted: 05/13/2019] [Indexed: 12/26/2022] Open
Abstract
In adult brain, the chemokine CXCL12 and its receptors CXCR4 and CXCR7 are expressed in neural progenitor and glial cells. Conditional Cxcl12 or Cxcr4 gene knockout in mice leads to severe alterations in neural progenitor proliferation, migration and differentiation. As adult hippocampal neurogenesis is involved in learning and memory processes, we investigated the long-term effects of reduced expression of CXCL12 or CXCR7 in heterozygous Cxcl12+/- and Cxcr7+/- animals (KD mice) on hippocampal neurogenesis, neuronal differentiation and memory processing. In Cxcl12 KD mice, Cxcr4 mRNA expression was reduced, whereas Cxcr7 was slightly increased. Conversely, in Cxcr7 KD mice, both Cxcr4 and Cxcl12 mRNA levels were decreased. Moreover, Cxcl12 KD animals showed marked behavioral and learning deficits that were associated with impaired neurogenesis in the hippocampus. Conversely, Cxcr7 KD animals showed mild learning deficits with normal neurogenesis, but reduced cell differentiation, measured with doublecortin immunolabeling. These findings suggested that a single Cxcl12 or Cxcr7 allele might not be sufficient to maintain the hippocampal niche functionality throughout life, and that heterozygosity might represent a susceptibility factor for memory dysfunction progression.
Collapse
|
31
|
Maurice T, Volle JN, Strehaiano M, Crouzier L, Pereira C, Kaloyanov N, Virieux D, Pirat JL. Neuroprotection in non-transgenic and transgenic mouse models of Alzheimer's disease by positive modulation of σ 1 receptors. Pharmacol Res 2019; 144:315-330. [PMID: 31048034 DOI: 10.1016/j.phrs.2019.04.026] [Citation(s) in RCA: 22] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2018] [Revised: 02/28/2019] [Accepted: 04/21/2019] [Indexed: 01/30/2023]
Abstract
The sigma-1 (σ1) receptor is an endoplasmic reticulum (ER) chaperone protein, enriched in mitochondria-associated membranes. Its activation triggers physiological responses to ER stress and modulate Ca2+ mobilization in mitochondria. Small σ1 agonist molecules activate the protein and act behaviorally as antidepressant, anti-amnesic and neuroprotective agents. Recently, several chemically unrelated molecules were shown to be σ1 receptor positive modulators (PMs), with some of them a clear demonstration of their allostericity. We here examined whether a σ1 PM also shows neuroprotective potentials in pharmacological and genetic models of Alzheimer's disease (AD). For this aim, we describe (±)-2-(3-chlorophenyl)-3,3,5,5-tetramethyl-2-oxo-[1,4,2]-oxazaphosphinane (OZP002) as a novel σ1 PM. OZP002 does not bind σ1 sites but induces σ1 effects in vivo and boosts σ1 agonist activity. OZP002 was antidepressant in the forced swim test and its effect was blocked by the σ1 antagonist NE-100 or in σ1 receptor knockout mice. It potentiated the antidepressant effect of the σ1 agonist igmesine. In mice tested for Y-maze alternation or passive avoidance, OZP002 prevented scopolamine-induced learning deficits, in a NE-100 sensitive manner. Pre-administered IP before an ICV injection of amyloid Aβ25-35 peptide, a pharmacological model of Alzheimer's disease, OZP002 prevented the learning deficits induced by the peptide after one week in the Y-maze, passive avoidance and novel object tests. Biochemical analyses of the mouse hippocampi showed that OZP002 significantly decreased Aβ25-35-induced increases in reactive oxygen species, lipid peroxidation, and increases in Bax, TNFα and IL-6 levels. Immunohistochemically, OZP002 prevented Aβ25-35-induced reactive astrogliosis and microgliosis in the hippocampus. It also alleviated Aβ25-35-induced decreases in synaptophysin level and choline acetyltransferase activity. Moreover, chronically administered in APPswe mice during 2 months, OZP002 prevented learning deficits (in all tests plus place learning in the water-maze) and increased biochemical markers. This study shows that σ1 PM with high neuropotective potential can be identified, combining pharmacological efficacy, selectivity and therapeutic safety, and identifies a novel promising compound, OZP002.
Collapse
Affiliation(s)
- Tangui Maurice
- MMDN, Univ Montpellier, INSERM, EPHE, UMR-S1198, Montpellier, France.
| | - Jean-Noël Volle
- Institut Charles Gerhardt, ENSCM, CNRS, UMR5253, Montpellier, France.
| | - Manon Strehaiano
- MMDN, Univ Montpellier, INSERM, EPHE, UMR-S1198, Montpellier, France.
| | - Lucie Crouzier
- MMDN, Univ Montpellier, INSERM, EPHE, UMR-S1198, Montpellier, France.
| | - Claire Pereira
- MMDN, Univ Montpellier, INSERM, EPHE, UMR-S1198, Montpellier, France.
| | - Nikolay Kaloyanov
- Institut Charles Gerhardt, ENSCM, CNRS, UMR5253, Montpellier, France.
| | - David Virieux
- Institut Charles Gerhardt, ENSCM, CNRS, UMR5253, Montpellier, France.
| | - Jean-Luc Pirat
- Institut Charles Gerhardt, ENSCM, CNRS, UMR5253, Montpellier, France.
| |
Collapse
|
32
|
Jadavji NM, Murray LK, Emmerson JT, Rudyk CA, Hayley S, Smith PD. Paraquat Exposure Increases Oxidative Stress Within the Dorsal Striatum of Male Mice With a Genetic Deficiency in One-carbon Metabolism. Toxicol Sci 2019; 169:25-33. [PMID: 30726997 PMCID: PMC6484892 DOI: 10.1093/toxsci/kfz034] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022] Open
Abstract
Paraquat is an herbicide that is commonly used worldwide. Exposure to paraquat results in Parkinson's disease (PD)-like symptoms including dopaminergic cell loss. Nutrition has also been linked in the pathogenesis of PD, such as reduced levels of folic acid, a B-vitamin, and component of one-carbon metabolism. Within one-carbon metabolism, methylenetetrahydrofolate reductase (MTHFR) catalyzes the irreversible conversion of 5, 10-methylenetetrahydrofolate to 5-methyltetrahydrofolate. A polymorphism in MTHFR (677 C&→T) has been reported in 5%-15% of North American and European human populations. The MTHFR polymorphism is also prevalent in PD patients. The goal of this study was to investigate the impact of paraquat-induced PD-like pathology in the context of reduced levels of MTHFR. Three-month-old male Mthfr+/- mice, which model the MTHFR polymorphism observed in humans, were administered intraperitoneal injections of paraquat (10 mg/kg) or saline 6 times over 3 weeks. At the end of paraquat treatment, motor and memory function were assessed followed by collection of brain tissue for biochemical analysis. Mthfr+/- mice treated with paraquat showed impaired motor function. There was increased microglial activation within the substantia nigra (SN) of Mthfr+/- mice treated with paraquat. Additionally, all Mthfr+/- mice that were treated with paraquat showed increased oxidative stress within the dorsal striatum, but not the SN. The present results show that paraquat exposure increases PD-like pathology in mice deficient in one-carbon metabolism.
Collapse
Affiliation(s)
- Nafisa M Jadavji
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Lauren K Murray
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Joshua T Emmerson
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Chris A Rudyk
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Shawn Hayley
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| | - Patrice D Smith
- Department of Neuroscience, Carleton University, Ottawa, Ontario, Canada
| |
Collapse
|
33
|
Theobromine Improves Working Memory by Activating the CaMKII/CREB/BDNF Pathway in Rats. Nutrients 2019; 11:nu11040888. [PMID: 31010016 PMCID: PMC6520707 DOI: 10.3390/nu11040888] [Citation(s) in RCA: 30] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2019] [Revised: 04/05/2019] [Accepted: 04/17/2019] [Indexed: 01/08/2023] Open
Abstract
Theobromine (TB) is a primary methylxanthine found in cacao beans. cAMP-response element-binding protein (CREB) is a transcription factor, which is involved in different brain processes that bring about cellular changes in response to discrete sets of instructions, including the induction of brain-derived neurotropic factor (BDNF). Ca2+/calmodulin-dependent protein kinase II (CaMKII) has been strongly implicated in the memory formation of different species as a key regulator of gene expression. Here we investigated whether TB acts on the CaMKII/CREB/BDNF pathway in a way that might improve the cognitive and learning function in rats. Male Wistar rats (5 weeks old) were divided into two groups. For 73 days, the control rats (CN rats) were fed a normal diet, while the TB-fed rats (TB rats) received the same food, but with a 0.05% TB supplement. To assess the effects of TB on cognitive and learning ability in rats: The radial arm maze task, novel object recognition test, and Y-maze test were used. Then, the brain was removed and the medial prefrontal cortex (mPFC) was isolated for Western Blot, real-time PCR and enzyme-linked immunosorbent assay. Phosphorylated CaMKII (p-CaMKII), phosphorylated CREB (p-CREB), and BDNF level in the mPFC were measured. In all the behavior tests, working memory seemed to be improved by TB ingestion. In addition, p-CaMKII and p-CREB levels were significantly elevated in the mPFC of TB rats in comparison to those of CN rats. We also found that cortical BDNF protein and mRNA levels in TB rats were significantly greater than those in CN rats. These results suggest that orally supplemented TB upregulates the CaMKII/CREB/BDNF pathway in the mPFC, which may then improve working memory in rats.
Collapse
|
34
|
Vavers E, Zvejniece L, Maurice T, Dambrova M. Allosteric Modulators of Sigma-1 Receptor: A Review. Front Pharmacol 2019; 10:223. [PMID: 30941035 PMCID: PMC6433746 DOI: 10.3389/fphar.2019.00223] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2018] [Accepted: 02/22/2019] [Indexed: 11/13/2022] Open
Abstract
Allosteric modulators of sigma-1 receptor (Sig1R) are described as compounds that can increase the activity of some Sig1R ligands that compete with (+)-pentazocine, one of the classic prototypical ligands that binds to the orthosteric Sig1R binding site. Sig1R is an endoplasmic reticulum membrane protein that, in addition to its promiscuous high-affinity ligand binding, has been shown to have chaperone activity. Different experimental approaches have been used to describe and validate the activity of allosteric modulators of Sig1R. Sig1R-modulatory activity was first found for phenytoin, an anticonvulsant drug that primarily acts by blocking the voltage-gated sodium channels. Accumulating evidence suggests that allosteric Sig1R modulators affect processes involved in the pathophysiology of depression, memory and cognition disorders as well as convulsions. This review will focus on the description of selective and non-selective allosteric modulators of Sig1R, including molecular structure properties and pharmacological activity both in vitro and in vivo, with the aim of providing the latest overview from compound discovery approaches to eventual clinical applications. In this review, the possible mechanisms of action will be discussed, and future challenges in the development of novel compounds will be addressed.
Collapse
Affiliation(s)
- Edijs Vavers
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Liga Zvejniece
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| | - Tangui Maurice
- MMDN, University of Montpellier, INSERM, EPHE, UMR-S1198, Montpellier, France
| | - Maija Dambrova
- Laboratory of Pharmaceutical Pharmacology, Latvian Institute of Organic Synthesis, Riga, Latvia
| |
Collapse
|
35
|
Prieur EAK, Jadavji NM. Assessing Spatial Working Memory Using the Spontaneous Alternation Y-maze Test in Aged Male Mice. Bio Protoc 2019; 9:e3162. [PMID: 33654968 DOI: 10.21769/bioprotoc.3162] [Citation(s) in RCA: 66] [Impact Index Per Article: 13.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2018] [Revised: 01/15/2019] [Accepted: 01/17/2019] [Indexed: 01/17/2023] Open
Abstract
The global population is aging and the prevalence of age-related diseases, such as Alzheimer's disease and vascular dementia is increasing. Understanding functional impairments and disease processes is of vital importance in order to develop effective therapeutics. Using the natural exploratory behavior of mice, the spontaneous alternation y-maze can assess short-term spatial working memory. The protocol for y-maze testing is straightforward and requires minimal resources, as well as animal training and output. Therefore, it can be broadly applied to study short-term memory in aged rodent models.
Collapse
|
36
|
Lalut J, Santoni G, Karila D, Lecoutey C, Davis A, Nachon F, Silman I, Sussman J, Weik M, Maurice T, Dallemagne P, Rochais C. Novel multitarget-directed ligands targeting acetylcholinesterase and σ 1 receptors as lead compounds for treatment of Alzheimer's disease: Synthesis, evaluation, and structural characterization of their complexes with acetylcholinesterase. Eur J Med Chem 2018; 162:234-248. [PMID: 30447434 DOI: 10.1016/j.ejmech.2018.10.064] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2018] [Revised: 10/24/2018] [Accepted: 10/29/2018] [Indexed: 12/19/2022]
Abstract
Pleiotropic intervention may be a requirement for effective limitation of the progression of multifactorial diseases such as Alzheimer's Disease. One approach to such intervention is to design a single chemical entity capable of acting on two or more targets of interest, which are accordingly known as Multi-Target Directed Ligands (MTDLs). We recently described donecopride, the first MTDL able to simultaneously inhibit acetylcholinesterase and act as an agonist of the 5-HT4 receptor, which displays promising activities in vivo. Pharmacomodulation of donecopride allowed us to develop a novel series of indole derivatives possessing interesting in vitro activities toward AChE and the σ1 receptor. The crystal structures of complexes of the most promising compounds with Torpedo californica AChE were solved in order to further understand their mode of inhibition.
Collapse
Affiliation(s)
- Julien Lalut
- Centre d'Etudes et de Recherche sur le Médicament de Normandie, Normandie Univ, UNICAEN, CERMN, 14000, Caen, France
| | - Gianluca Santoni
- Univ Grenoble Alpes, IBS, F-38027, Grenoble, France; CNRS, IBS, F-38027, Grenoble, France; CEA, IBS, F-38027, Grenoble, France
| | - Delphine Karila
- Centre d'Etudes et de Recherche sur le Médicament de Normandie, Normandie Univ, UNICAEN, CERMN, 14000, Caen, France
| | - Cédric Lecoutey
- Centre d'Etudes et de Recherche sur le Médicament de Normandie, Normandie Univ, UNICAEN, CERMN, 14000, Caen, France
| | - Audrey Davis
- Centre d'Etudes et de Recherche sur le Médicament de Normandie, Normandie Univ, UNICAEN, CERMN, 14000, Caen, France
| | - Florian Nachon
- Département de Toxicologie et Risques Chimiques, Institut de Recherche Biomédicale des Armées, 91220, Brétigny-sur-Orge, France
| | - Israel Silman
- Department of Neurobiology, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Joel Sussman
- Department of Structural Biology, Weizmann Institute of Science, 76100, Rehovot, Israel
| | - Martin Weik
- Univ Grenoble Alpes, IBS, F-38027, Grenoble, France; CNRS, IBS, F-38027, Grenoble, France; CEA, IBS, F-38027, Grenoble, France
| | - Tangui Maurice
- Molecular Mechanisms in Neurodegenerative Diseases, MMDN Laboratory, University of Montpellier, Ecole Pratique des Hautes Etudes, Institut National de la Recherche et de la Santé Médicale, UMR-S1198, 34095, Montpellier, France
| | - Patrick Dallemagne
- Centre d'Etudes et de Recherche sur le Médicament de Normandie, Normandie Univ, UNICAEN, CERMN, 14000, Caen, France.
| | - Christophe Rochais
- Centre d'Etudes et de Recherche sur le Médicament de Normandie, Normandie Univ, UNICAEN, CERMN, 14000, Caen, France.
| |
Collapse
|
37
|
Chen Y, Milenkovic M, Horsfall W, Salahpour A, Soderling SH, Ramsey AJ. Restoring striatal WAVE-1 improves maze exploration performance of GluN1 knockdown mice. PLoS One 2018; 13:e0199341. [PMID: 30352064 PMCID: PMC6198945 DOI: 10.1371/journal.pone.0199341] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2018] [Accepted: 10/06/2018] [Indexed: 11/19/2022] Open
Abstract
NMDA receptors are important for cognition and are implicated in neuropsychiatric disorders. GluN1 knockdown (GluN1KD) mice have reduced NMDA receptor levels, striatal spine density deficits, and cognitive impairments. However, how NMDA depletion leads to these effects is unclear. Since Rho GTPases are known to regulate spine density and cognition, we examined the levels of RhoA, Rac1, and Cdc42 signaling proteins. Striatal Rac1-pathway components are reduced in GluN1KD mice, with Rac1 and WAVE-1 deficits at 6 and 12 weeks of age. Concurrently, medium spiny neuron (MSN) spine density deficits are present in mice at these ages. To determine whether WAVE-1 deficits were causal or compensatory in relation to these phenotypes, we intercrossed GluN1KD mice with WAVE-1 overexpressing (WAVE-Tg) mice to restore WAVE-1 levels. GluN1KD-WAVE-Tg hybrids showed rescue of striatal WAVE-1 protein levels and MSN spine density, as well as selective behavioral rescue in the Y-maze and 8-arm radial maze tests. GluN1KD-WAVE-Tg mice expressed normalized WAVE-1 protein levels in the hippocampus, yet spine density of hippocampal CA1 pyramidal neurons was not significantly altered. Our data suggest a nuanced role for WAVE-1 effects on cognition and a delineation of specific cognitive domains served by the striatum. Rescue of striatal WAVE-1 and MSN spine density may be significant for goal-directed exploration and associated long-term memory in mice.
Collapse
Affiliation(s)
- Yuxiao Chen
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Marija Milenkovic
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Wendy Horsfall
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Ali Salahpour
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| | - Scott H. Soderling
- Department of Cell Biology, Duke University Medical Center, Durham, NC, United States of America
| | - Amy J. Ramsey
- Department of Pharmacology and Toxicology, University of Toronto, Toronto, ON, Canada
| |
Collapse
|
38
|
Maurice T, Strehaiano M, Duhr F, Chevallier N. Amyloid toxicity is enhanced after pharmacological or genetic invalidation of the σ 1 receptor. Behav Brain Res 2018; 339:1-10. [PMID: 29129596 DOI: 10.1016/j.bbr.2017.11.010] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Accepted: 11/07/2017] [Indexed: 10/18/2022]
Abstract
The sigma-1 receptor (S1R) is a molecular chaperone which activity modulates several intracellular signals including calcium mobilization at mitochondria-associated endoplasmic reticulum membranes. S1R agonists are potent neuroprotectants against neurodegenerative insults and particularly in rodent models of Alzheimer's disease (AD). We here analyzed whether S1R inactivation modifies vulnerability to amyloid toxicity in AD models. Two strategies were used: (1) amyloid β[25-35] (Aβ25-35) peptide (1, 3, 9nmol) was injected intracerebroventricularly in mice treated repeatedly with the S1R antagonist NE-100 or in S1RKO mice, and (2) WT, APPSweInd, S1RKO, and APPSweInd/S1RKO mice were created and female littermates analyzed at 8 months of age. Learning deficits, oxidative stress, Bax level and BDNF content in the hippocampus were analyzed. Aβ25-35 induced learning impairment, oxidative stress, Bax induction and BDNF alteration at lower dose in NE-100-treated mice or S1RKO mice as compared to WT animals. The extent of learning deficits and biochemical alterations were also higher in APPSweInd/S1RKO mice as compared to WT, APPSweInd, and S1RKO animals. S1R inactivation or altered S1R expression augmented the pathological status in pharmacologic and genetic AD mouse models. These observations, in relation with the well-known protective effects of S1R agonists, are coherent with a role of signal amplifier in neurodegeneration and neuroprotection proposed for S1R in AD and related neurodegenerative disorders.
Collapse
Affiliation(s)
- Tangui Maurice
- MMDN, Univ. Montpellier, EPHE, INSERM, UMR-S1198, Montpellier, F-34095, France.
| | - Manon Strehaiano
- MMDN, Univ. Montpellier, EPHE, INSERM, UMR-S1198, Montpellier, F-34095, France
| | - Fanny Duhr
- MMDN, Univ. Montpellier, EPHE, INSERM, UMR-S1198, Montpellier, F-34095, France
| | - Nathalie Chevallier
- MMDN, Univ. Montpellier, EPHE, INSERM, UMR-S1198, Montpellier, F-34095, France
| |
Collapse
|
39
|
Prieur EAK, Pjetri E, Zeisel SH, Jadavji NM. Reduced brain volume and impaired memory in betaine homocysteine S-methyltransferase knockout mice. Appl Physiol Nutr Metab 2017; 42:1228-1231. [PMID: 28715642 DOI: 10.1139/apnm-2017-0182] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Using a mouse model, this study examined the impact of lack of betaine homocysteine S-methyltransferase (BHMT) on neurological function. Bhmt-/- mice maintained on a control diet had elevated concentrations of homocysteine, reduced total brain magnetic resonance imaging (MRI) volume, as well as impaired reference and short-term memories. The results of this study indicate that the absence of BHMT may play a role in neurological function.
Collapse
Affiliation(s)
- Emily A K Prieur
- a Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada
| | - Eneda Pjetri
- b Department of Nutrition, UNC Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599-7461, USA.,c UNC School of Medicine, University of North Carolina, Chapel Hill, NC 27516, USA.,d Nutrition Research Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 28081, USA
| | - Steven H Zeisel
- b Department of Nutrition, UNC Gillings School of Global Public Health, University of North Carolina, Chapel Hill, NC 27599-7461, USA.,c UNC School of Medicine, University of North Carolina, Chapel Hill, NC 27516, USA.,d Nutrition Research Institute, University of North Carolina at Chapel Hill, Chapel Hill, NC 28081, USA
| | - Nafisa M Jadavji
- a Department of Neuroscience, Carleton University, 1125 Colonel By Drive, Ottawa, ON K1S 5B6, Canada.,e Department of Experimental Neurology, Center for Stroke Research Berlin, Charité University Medicine Berlin, Berlin 10117, Germany
| |
Collapse
|
40
|
Muradashvili N, Tyagi SC, Lominadze D. Localization of Fibrinogen in the Vasculo-Astrocyte Interface after Cortical Contusion Injury in Mice. Brain Sci 2017; 7:brainsci7070077. [PMID: 28684673 PMCID: PMC5532590 DOI: 10.3390/brainsci7070077] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2017] [Revised: 06/30/2017] [Accepted: 07/04/2017] [Indexed: 01/27/2023] Open
Abstract
Besides causing neuronal damage, traumatic brain injury (TBI) is involved in memory reduction, which can be a result of alterations in vasculo-neuronal interactions. Inflammation following TBI is involved in elevation of blood content of fibrinogen (Fg), which is known to enhance cerebrovascular permeability, and thus, enhance its deposition in extravascular space. However, the localization of Fg in the extravascular space and its possible interaction with nonvascular cells are not clear. The localization of Fg deposition in the extravascular space was defined in brain samples of mice after cortical contusion injury (CCI) and sham-operation (control) using immunohistochemistry and laser-scanning confocal microscopy. Memory changes were assessed with new object recognition and Y-maze tests. Data showed a greater deposition of Fg in the vascular and astrocyte endfeet interface in mice with CCI than in control animals. This effect was accompanied by enhanced neuronal degeneration and reduction in short-term memory in mice with CCI. Thus, our results suggest that CCI induces increased deposition of Fg in the vasculo-astrocyte interface, and is accompanied by neuronal degeneration, which may result in reduction of short-term memory.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY 40202, USA.
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY 40202, USA.
| | - David Lominadze
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY 40202, USA.
| |
Collapse
|
41
|
Kubo KI, Deguchi K, Nagai T, Ito Y, Yoshida K, Endo T, Benner S, Shan W, Kitazawa A, Aramaki M, Ishii K, Shin M, Matsunaga Y, Hayashi K, Kakeyama M, Tohyama C, Tanaka KF, Tanaka K, Takashima S, Nakayama M, Itoh M, Hirata Y, Antalffy B, Armstrong DD, Yamada K, Inoue K, Nakajima K. Association of impaired neuronal migration with cognitive deficits in extremely preterm infants. JCI Insight 2017; 2:88609. [PMID: 28515367 DOI: 10.1172/jci.insight.88609] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/16/2016] [Accepted: 04/18/2017] [Indexed: 12/23/2022] Open
Abstract
Many extremely preterm infants (born before 28 gestational weeks [GWs]) develop cognitive impairment in later life, although the underlying pathogenesis is not yet completely understood. Our examinations of the developing human neocortex confirmed that neuronal migration continues beyond 23 GWs, the gestational week at which extremely preterm infants have live births. We observed larger numbers of ectopic neurons in the white matter of the neocortex in human extremely preterm infants with brain injury and hypothesized that altered neuronal migration may be associated with cognitive impairment in later life. To confirm whether preterm brain injury affects neuronal migration, we produced brain damage in mouse embryos by occluding the maternal uterine arteries. The mice showed delayed neuronal migration, ectopic neurons in the white matter, altered neuronal alignment, and abnormal corticocortical axonal wiring. Similar to human extremely preterm infants with brain injury, the surviving mice exhibited cognitive deficits. Activation of the affected medial prefrontal cortices of the surviving mice improved working memory deficits, indicating that decreased neuronal activity caused the cognitive deficits. These findings suggest that altered neuronal migration altered by brain injury might contribute to the subsequent development of cognitive impairment in extremely preterm infants.
Collapse
Affiliation(s)
- Ken-Ichiro Kubo
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Kimiko Deguchi
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan.,Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan.,Department of Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - Taku Nagai
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Yukiko Ito
- Department of Molecular Neuroscience, Medical Research Institute/School of Biomedical Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Keitaro Yoshida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Toshihiro Endo
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Seico Benner
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan
| | - Wei Shan
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ayako Kitazawa
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan.,Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Michihiko Aramaki
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Kazuhiro Ishii
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Minkyung Shin
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Yuki Matsunaga
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Kanehiro Hayashi
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Masaki Kakeyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.,Laboratory for Systems Neuroscience & Preventive Medicine, Waseda University Faculty of Human Sciences, Tokorozawa, Japan
| | - Chiharu Tohyama
- Laboratory of Environmental Health Sciences, Center for Disease Biology and Integrative Medicine, Graduate School of Medicine, University of Tokyo, Tokyo, Japan.,Environmental Biology Laboratory, Faculty of Medicine, University of Tsukuba, Tsukuba, Japan
| | - Kenji F Tanaka
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Kohichi Tanaka
- Department of Molecular Neuroscience, Medical Research Institute/School of Biomedical Science, Tokyo Medical and Dental University, Tokyo, Japan
| | - Sachio Takashima
- Division of Child Neurology, Yanagawa Institute of Developmental Disabilities, Yanagawa, Japan
| | - Masahiro Nakayama
- Department of Pathology, Osaka Medical Center and Research Institute for Maternal and Child Health, Izumi, Japan
| | - Masayuki Itoh
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Yukio Hirata
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| | - Barbara Antalffy
- Department of Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - Dawna D Armstrong
- Department of Pathology, Baylor College of Medicine, Houston, Texas, USA
| | - Kiyofumi Yamada
- Department of Neuropsychopharmacology and Hospital Pharmacy, Nagoya University Graduate School of Medicine, Nagoya, Japan
| | - Ken Inoue
- Department of Mental Retardation and Birth Defect Research, National Institute of Neuroscience, National Center of Neurology and Psychiatry, Kodaira, Japan
| | - Kazunori Nakajima
- Department of Anatomy, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
42
|
Dam K, Füchtemeier M, Farr TD, Boehm-Sturm P, Foddis M, Dirnagl U, Malysheva O, Caudill MA, Jadavji NM. Increased homocysteine levels impair reference memory and reduce cortical levels of acetylcholine in a mouse model of vascular cognitive impairment. Behav Brain Res 2017; 321:201-208. [DOI: 10.1016/j.bbr.2016.12.041] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/27/2016] [Revised: 12/26/2016] [Accepted: 12/29/2016] [Indexed: 01/26/2023]
|
43
|
Maurice T, Goguadze N. Role of σ 1 Receptors in Learning and Memory and Alzheimer's Disease-Type Dementia. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:213-233. [PMID: 28315274 DOI: 10.1007/978-3-319-50174-1_15] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The present chapter will review the role of σ1 receptor in learning and memory and neuroprotection , against Alzheimer's type dementia. σ1 Receptor agonists have been tested in a variety of pharmacological and pathological models of learning impairments in rodents these last past 20 years. Their anti-amnesic effects have been explained by the wide-range modulatory role of σ1 receptors on Ca2+ mobilizations, neurotransmitter responses, and particularly glutamate and acetylcholine systems, and neurotrophic factors. Recent observations from genetic and pharmacological studies have shown that σ1 receptor can also be targeted in neurodegenerative diseases, and particularly Alzheimer's disease . Several compounds, acting partly through the σ1 receptor, have showed effective neuroprotection in transgenic mouse models of Alzheimer's disease . We will review the data and discuss the possible mechanisms of action, particularly focusing on oxidative stress and mitochondrial integrity, trophic factors and a novel hypothesis suggesting a functional interaction between the σ1 receptor and α7 nicotinic acetylcholine receptor. Finally, we will discuss the pharmacological peculiarities of non-selective σ1 receptor ligands, now developed as neuroprotectants in Alzheimer's disease , and positive modulators, recently described and that showed efficacy against learning and memory deficits.
Collapse
Affiliation(s)
- Tangui Maurice
- INSERM U1198, University of Montpellier, 34095, Montpellier, France.
| | - Nino Goguadze
- INSERM U1198, University of Montpellier, 34095, Montpellier, France
- Institute of Chemical Biology, Ilia State University, Tbilisi, 0162, GA, USA
| |
Collapse
|
44
|
Abstract
Sigma1 (also known as sigma-1 receptor, Sig1R, σ1 receptor) is a unique pharmacologically regulated integral membrane chaperone or scaffolding protein. The majority of publications on the subject have focused on the neuropharmacology of Sigma1. However, a number of publications have also suggested a role for Sigma1 in cancer. Although there is currently no clinically used anti-cancer drug that targets Sigma1, a growing body of evidence supports the potential of Sigma1 ligands as therapeutic agents to treat cancer. In preclinical models, compounds with affinity for Sigma1 have been reported to inhibit cancer cell proliferation and survival, cell adhesion and migration, tumor growth, to alleviate cancer-associated pain, and to have immunomodulatory properties. This review will highlight that although the literature supports a role for Sigma1 in cancer, several fundamental questions regarding drug mechanism of action and the physiological relevance of aberrant SIGMAR1 transcript and Sigma1 protein expression in certain cancers remain unanswered or only partially answered. However, emerging lines of evidence suggest that Sigma1 is a component of the cancer cell support machinery, that it facilitates protein interaction networks, that it allosterically modulates the activity of its associated proteins, and that Sigma1 is a selectively multifunctional drug target.
Collapse
Affiliation(s)
- Felix J Kim
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, USA.
- Sidney Kimmel Cancer Center, Philadelphia, PA, USA.
| | - Christina M Maher
- Department of Pharmacology and Physiology, Drexel University College of Medicine, 245 North 15th Street, Philadelphia, PA, USA
| |
Collapse
|
45
|
Deng M, Huang L, Ning B, Wang N, Zhang Q, Zhu C, Fang Y. β-asarone improves learning and memory and reduces Acetyl Cholinesterase and Beta-amyloid 42 levels in APP/PS1 transgenic mice by regulating Beclin-1-dependent autophagy. Brain Res 2016; 1652:188-194. [PMID: 27737765 DOI: 10.1016/j.brainres.2016.10.008] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/20/2016] [Revised: 10/07/2016] [Accepted: 10/09/2016] [Indexed: 12/11/2022]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder in the elderly, and studies have suggested that β-asarone has pharmacological effects on beta-amyloid (Aβ) injected in the rat hippocampus. However, the effect of β-asarone on autophagy in the APP/PS1 transgenic mouse is unreported. APP/PS1 transgenic mice were randomly divided into six groups (n=10/group): an untreated group, an Aricept-treated group, a 3-MA-treated group, a rapamycin-treated group, an LY294002-treated group, a β-asarone-treated group. The control group consisted of wild-type C57BL/6 mice. All treatments were administered to the mice for 30 days. Spatial learning and memory were assessed by water maze, passive avoidance, and step-down tests. AChE and Aβ42 levels in the hippocampus were determined by ELISA. p-Akt, p-mTOR, and LC3B expression were detected by flow cytometry. The expression of p-Akt, p-mTOR, Beclin-1, and p62 proteins was assessed by western blot. Changes in autophagy were viewed using a transmission electron microscope. APP and Beclin-1 mRNA levels were measured by Real-Time PCR. The learning and memory of APP/PS1 transgenic mice were improved significantly after β-asarone treatment compared with the untreated group. In addition, β-asarone treatment reduced AChE and Aβ42 levels, increased p-mTOR and p62 expression, decreased p-Akt, Beclin-1, and LC3B expression, decreased the number of autophagosomes and reduced APP mRNA and Beclin-1 mRNA levels compared with the untreated group. That is, β-asarone treatment can improve the learning and memory abilities of APP/PS1 transgenic mouse by inhibiting Beclin-1-dependent autophagy via the PI3K/Akt/mTOR pathway.
Collapse
Affiliation(s)
- Minzhen Deng
- Guangdong Provincial Hospital of Chinese Medicine, Guangdong Provincial Academy of Chinese Medical Sciences, and The Second Clinical College, Guangzhou University of Chinese Medicine, Guangzhou 510120, PR China; Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Liping Huang
- Hainan Medical University, Haikou 571199, PR China; Lingnan Normal University, Zhanjiang 524048, PR China
| | - Baile Ning
- Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Nanbu Wang
- Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Qinxin Zhang
- Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Caixia Zhu
- Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China
| | - Yongqi Fang
- Guangzhou University of Chinese Medicine, Guangzhou 510006, PR China; The First Affiliated Hospital of Guangzhou University of Chinese Medicine, Guangzhou 510405, PR China.
| |
Collapse
|
46
|
Abdel-Hafiz L, Chao OY, Huston JP, Nikolaus S, Spieler RE, de Souza Silva MA, Mattern C. Promnestic effects of intranasally applied pregnenolone in rats. Neurobiol Learn Mem 2016; 133:185-195. [DOI: 10.1016/j.nlm.2016.07.012] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2016] [Revised: 06/24/2016] [Accepted: 07/12/2016] [Indexed: 10/21/2022]
|
47
|
Muradashvili N, Tyagi R, Tyagi N, Tyagi SC, Lominadze D. Cerebrovascular disorders caused by hyperfibrinogenaemia. J Physiol 2016; 594:5941-5957. [PMID: 27121987 DOI: 10.1113/jp272558] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Accepted: 04/25/2016] [Indexed: 01/18/2023] Open
Abstract
KEY POINTS Hyperfibrinogenaemia (HFg) results in vascular remodelling, and fibrinogen (Fg) and amyloid β (Aβ) complex formation is a hallmark of Alzheimer's disease. However, the interconnection of these effects, their mechanisms and implications in cerebrovascular diseases are not known. Using a mouse model of HFg, we showed that at an elevated blood level, Fg increases cerebrovascular permeability via mainly caveolar protein transcytosis. This enhances deposition of Fg in subendothelial matrix and interstitium making the immobilized Fg a readily accessible substrate for binding Aβ and cellular prion protein (PrPC ), the protein that is thought to have a greater effect on memory than Aβ. We showed that enhanced formation of Fg-Aβ and Fg-PrPC complexes are associated with reduction in short-term memory. The present study delineates a new mechanistic pathway for vasculo-neuronal dysfunctions found in inflammatory cardiovascular and cerebrovascular diseases associated with an elevated blood level of Fg. ABSTRACT Many cardiovascular diseases are associated with inflammation and as such are accompanied by an increased blood level of fibrinogen (Fg). Besides its well-known prothrombotic effects Fg seems to have other destructive roles in developing microvascular dysfunction that include changes in vascular reactivity and permeability. Increased permeability of brain microvessels has the most profound effects as it may lead to cerebrovascular remodelling and result in memory reduction. The goal of the present study was to define mechanisms of cerebrovascular permeability and associated reduction in memory induced by elevated blood content of Fg. Genetically modified, transgenic hyperfibrinogenic (HFg) mice were used to study cerebrovascular transcellular and paracellular permeability in vivo. The extent of caveolar formation and the role of caveolin-1 signalling were evaluated by immunohistochemistry (IHC) and Western blot (WB) analysis in brain samples from experimental animals. Formation of Fg complexes with amyloid β (Aβ) and with cellular prion protein (PrPC ) were also assessed with IHC and WB analysis. Short-term memory of mice was assessed by novel object recognition and Y-maze tests. Caveolar protein transcytosis was found to have a prevailing role in overall increased cerebrovascular permeability in HFg mice. These results were associated with enhanced formation of caveolae. Increased formation of Fg-PrPC and Fg-Aβ complexes were correlated with reduction in short-term memory in HFg mice. Using the model of hyperfibrinogenaemia, the present study shows a novel mechanistic pathway of inflammation-induced and Fg-mediated reduction in short-term memory.
Collapse
Affiliation(s)
- Nino Muradashvili
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Reeta Tyagi
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Neetu Tyagi
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - Suresh C Tyagi
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA
| | - David Lominadze
- Department of Physiology, University of Louisville, School of Medicine, Louisville, KY, USA.
| |
Collapse
|
48
|
AC-3933, a benzodiazepine partial inverse agonist, improves memory performance in MK-801-induced amnesia mouse model. Pharmacol Biochem Behav 2016; 144:45-52. [PMID: 26946254 DOI: 10.1016/j.pbb.2016.03.001] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/28/2015] [Revised: 02/06/2016] [Accepted: 03/01/2016] [Indexed: 01/28/2023]
Abstract
AC-3933, a novel benzodiazepine receptor partial inverse agonist, is a drug candidate for cognitive disorders including Alzheimer's disease. We have previously reported that AC-3933 enhances acetylcholine release in the rat hippocampus and ameliorates scopolamine-induced memory impairment and age-related cognitive decline in both rats and mice. In this study, we further evaluated the procognitive effect of AC-3933 on memory impairment induced by MK-801, an N-methyl-d-aspartate receptor antagonist, in mice. Unlike the acetylcholinesterase inhibitor donepezil and the benzodiazepine receptor inverse agonist FG-7142, oral administration of AC-3933 significantly ameliorated MK-801-induced memory impairment in the Y-maze test and in the object location test. Interestingly, the procognitive effects of AC-3933 on MK-801-induced memory impairment were not affected by the benzodiazepine receptor antagonist flumazenil, although this was not the case for the beneficial effects of AC-3933 on scopolamine-induced memory deficit. Moreover, the onset of AC-3933 ameliorating effect on scopolamine- or MK-801-induced memory impairment was different in the Y-maze test. Taken together, these results indicate that AC-3933 improves memory deficits caused by both cholinergic and glutamatergic hypofunction and suggest that the ameliorating effect of AC-3933 on MK-801-induced memory impairment is mediated by a mechanism other than inverse activation of the benzodiazepine receptor.
Collapse
|
49
|
Maurice T. Protection by sigma-1 receptor agonists is synergic with donepezil, but not with memantine, in a mouse model of amyloid-induced memory impairments. Behav Brain Res 2016; 296:270-278. [PMID: 26386305 DOI: 10.1016/j.bbr.2015.09.020] [Citation(s) in RCA: 48] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/11/2015] [Accepted: 09/15/2015] [Indexed: 12/14/2022]
Abstract
Drugs activating the sigma-1 (σ1) chaperone protein are anti-amnesic and neuroprotective in neurodegenerative pathologies like Alzheimer's disease (AD). Since these so-called σ1 receptor (σ1R) agonists modulate cholinergic and glutamatergic systems in a variety of physiological responses, we addressed their putative additive/synergistic action in combination with cholinergic or glutamatergic drugs. The selective σ1 agonist PRE-084, or the non-selective σ1 drug ANAVEX2-73 was combined with the acetylcholinesterase inhibitor donepezil or the NMDA receptor antagonist memantine in the nontransgenic mouse model of AD-like memory impairments induced by intracerebroventricular injection of oligomeric Aβ25-35 peptide. Two behavioral tests, spontaneous alternation and passive avoidance response, were used in parallel and both protective and symptomatic effects were examined. After determination of the minimally active doses for each compound, the combinations were tested and the combination index (CI) calculated. Combinations between the σ1 agonists and donepezil showed a synergic protective effect, with CI<1, whereas the combinations with memantine showed an antagonist effect, with CI>1. Symptomatic effects appeared only additive for all combinations, with CI=1. A pharmacological analysis of the PRE-084+donepezil combination revealed that the synergy could be due to an inter-related mechanism involving α7 nicotinic ACh receptors and σ1R. These results demonstrated that σ1 drugs do not only offer a protective potential alone but also in combination with other therapeutic agents. The nature of neuromodulatory molecular chaperone of the σ1R could eventually lead to synergistic combinations.
Collapse
Affiliation(s)
- Tangui Maurice
- Molecular Mechanisms in Neurodegenerative Diseases, MMDN Laboratory, Institut National de la Recherche et de la Santé Médicale, unit 1198, 34095 Montpellier, France; University of Montpellier, 34095 Montpellier, France; Ecole Pratique des Hautes Etudes, 75014 Paris, France.
| |
Collapse
|
50
|
Neuronal Heterotopias Affect the Activities of Distant Brain Areas and Lead to Behavioral Deficits. J Neurosci 2015; 35:12432-45. [PMID: 26354912 DOI: 10.1523/jneurosci.3648-14.2015] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Neuronal heterotopia refers to brain malformations resulting from deficits of neuronal migration. Individuals with heterotopias show a high incidence of neurological deficits, such as epilepsy. More recently, it has come to be recognized that focal heterotopias may also show a range of psychiatric problems, including cognitive and behavioral impairments. However, because focal heterotopias are not always located in the brain areas responsible for the symptoms, the causal relationship between the symptoms and heterotopias remains elusive. In this study, we showed that mice with focal heterotopias in the somatosensory cortex generated by in utero electroporation exhibited spatial working memory deficit and low competitive dominance behavior, which have been shown to be closely associated with the activity of the medial prefrontal cortex (mPFC) in rodents. Analysis of the mPFC activity revealed that the immediate-early gene expression was decreased and the local field potentials of the mPFC were altered in the mice with heterotopias compared with the control mice. Moreover, activation of these ectopic and overlying sister neurons using the DREADD (designer receptor exclusively activated by designer drug) system improved the working memory deficits. These findings suggest that cortical regions containing focal heterotopias can affect distant brain regions and give rise to behavioral abnormalities. Significance statement: Recent studies reported that patients with heterotopias have a variety of clinical symptoms, such as cognitive disturbance, psychiatric symptoms, and autistic behavior. However, the causal relationship between the symptoms and heterotopias remains elusive. Here we showed that mice with focal heterotopias in the somatosensory cortex generated by in utero electroporation exhibited behavioral deficits that have been shown to be associated with the mPFC activity in rodents. The existence of heterotopias indeed altered the neural activities of the mPFC, and direct manipulation of the neural activity of the ectopic neurons and their sister neurons in the overlying cortex improved the behavioral deficit. Thus, our results indicate that focal heterotopias could affect the activities of distant brain areas and cause behavioral abnormalities.
Collapse
|