1
|
Li Y, Dahl K, Johnström P, Varnäs K, Farde L, Halldin C, Medd A, Maier D, Powell ME, Chen J, Van R, Patel J, Chaudhary A, Gao Y, Song Z, Haider A, Shao Y, Elmore CS, Liang S, Schou M. Radiosynthesis and Evaluation of 11C-Labeled Isoindolone-Based Positive Allosteric Modulators for Positron Emission Tomography Imaging of Metabotropic Glutamate Receptor 2. ACS Pharmacol Transl Sci 2024; 7:2414-2423. [PMID: 39144551 PMCID: PMC11320742 DOI: 10.1021/acsptsci.4c00261] [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: 05/03/2024] [Revised: 06/23/2024] [Accepted: 06/26/2024] [Indexed: 08/16/2024]
Abstract
The metabotropic glutamate receptor 2 (mGluR2) has emerged as a potential therapeutic target for the treatment of various neurological diseases, prompting substantial interest in the development of mGluR2-targeted drug candidates. As part of our medicinal chemistry program, we synthesized a series of isoindolone derivatives and assessed their potential as mGluR2 positive allosteric modulators (PAMs). Notably, AZ12559322 exhibited high affinity (K i mGluR2 = 1.31 nM) and an excellent in vitro binding specificity of 89% while demonstrating selectivity over other mGluR subtypes (>4000-fold). Autoradiography with the radiolabeled counterpart, [3H]AZ12559322, revealed a heterogeneous accumulation with the highest binding in mGluR2-rich brain regions. Radioligand binding was significantly reduced by pretreatment with nonradioactive mGluR2 PAMs in brains of rats and nonhuman primates. Although positron emission tomography imaging of [11C]AZ12559322 (6a) revealed low brain uptake in a nonhuman primate, this study provides valuable guidance to further design novel isoindolone-based mGluR2 PAMs with improved brain exposure.
Collapse
Affiliation(s)
- Yinlong Li
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton
Road, Atlanta, Georgia 30322, United States
| | - Kenneth Dahl
- PET
Science Centre, Precision Medicine and Biosamples, Oncology R&D,
AstraZeneca, Karolinska Institutet, Stockholm S-17176, Sweden
- Department
of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm S-17176, Sweden
| | - Peter Johnström
- PET
Science Centre, Precision Medicine and Biosamples, Oncology R&D,
AstraZeneca, Karolinska Institutet, Stockholm S-17176, Sweden
- Department
of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm S-17176, Sweden
| | - Katarina Varnäs
- Department
of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm S-17176, Sweden
| | - Lars Farde
- Department
of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm S-17176, Sweden
| | - Christer Halldin
- Department
of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm S-17176, Sweden
| | - Amy Medd
- Neuroscience,
BioPharmaceuticals R&D, AstraZeneca, Wilmington, Delaware 19803, United States
| | - Donna Maier
- Neuroscience,
BioPharmaceuticals R&D, AstraZeneca, Wilmington, Delaware 19803, United States
| | - Mark E. Powell
- Neuroscience,
BioPharmaceuticals R&D, AstraZeneca, Wilmington, Delaware 19803, United States
| | - Jiahui Chen
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton
Road, Atlanta, Georgia 30322, United States
| | - Richard Van
- Department
of Chemistry and Biochemistry, University
of Oklahoma, Norman, Oklahoma 73019-5251, United States
| | - Jimmy Patel
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton
Road, Atlanta, Georgia 30322, United States
| | - Ahmad Chaudhary
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton
Road, Atlanta, Georgia 30322, United States
| | - Yabiao Gao
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton
Road, Atlanta, Georgia 30322, United States
| | - Zhendong Song
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton
Road, Atlanta, Georgia 30322, United States
| | - Ahmed Haider
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton
Road, Atlanta, Georgia 30322, United States
| | - Yihan Shao
- Department
of Chemistry and Biochemistry, University
of Oklahoma, Norman, Oklahoma 73019-5251, United States
| | - Charles S. Elmore
- Neuroscience,
BioPharmaceuticals R&D, AstraZeneca, Wilmington, Delaware 19803, United States
- Early
Chemical Development, Pharmaceutical Sciences, R&D, AstraZeneca Pharmaceuticals, Gothenburg 43183, Sweden
| | - Steven Liang
- Department
of Radiology and Imaging Sciences, Emory
University, 1364 Clifton
Road, Atlanta, Georgia 30322, United States
| | - Magnus Schou
- PET
Science Centre, Precision Medicine and Biosamples, Oncology R&D,
AstraZeneca, Karolinska Institutet, Stockholm S-17176, Sweden
- Department
of Clinical Neuroscience, Centre for Psychiatry Research, Karolinska Institutet and Stockholm County Council, Stockholm S-17176, Sweden
| |
Collapse
|
2
|
Kaar A, Weir MP, Rae MG. Altered neuronal group 1 metabotropic glutamate receptor- and endoplasmic reticulum-mediated Ca 2+ signaling in two rodent models of Alzheimer's disease. Neurosci Lett 2024; 823:137664. [PMID: 38309326 DOI: 10.1016/j.neulet.2024.137664] [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: 06/12/2023] [Revised: 01/15/2024] [Accepted: 01/29/2024] [Indexed: 02/05/2024]
Abstract
Calcium mobilization from the endoplasmic reticulum (ER) induced by, for example, IP3 receptor (IP3R) stimulation, and its subsequent crosstalk with extracellular Ca2+ influx mediated through voltage-gated calcium channels (VGCCs) and neuronal store-operated calcium entry (nSOCE), is essential for normal neuronal signaling and cellular homeostasis. However, several studies suggest that chronic calcium dysregulation may play a key role in the onset and/or progression of neurodegenerative conditions, particularly Alzheimer's disease (AD). Here, using early postnatal hippocampal tissue from two transgenic murine models of AD, we provide further evidence that not only are crucial calcium signaling pathways dysregulated, but also that such dysregulation occurs at very early stages of development. Utilizing epifluorescence calcium imaging, we investigated ER-, nSOCE- and VGCC-mediated calcium signaling in cultured primary hippocampal neurons from two transgenic rodent models of AD: 3xTg-AD mice (PS1M146V/APPSWE/TauP301L) and TgF344-AD rats (APPSWE/PS1ΔE9) between 2 and 9 days old. Our results reveal that, in comparison to control hippocampal neurons, those from 3xTg-AD mice possessed significantly greater basal ER calcium levels, as measured by larger responses to I-mGluR-mediated ER Ca2+ mobilization (amplitude; 4 (0-19) vs 21(12-36) a.u., non-Tg vs 3xTg-AD; median difference (95 % Cl) = 14 a.u. (11-18); p = 0.004)) but reduced nSOCE (15 (4-22) vs 8(5-11) a.u., non-Tg vs 3xTg-AD; median difference (95 % Cl) = -7 a.u. (-3- -10 a.u.); p < 0.0001). Furthermore, unlike non-Tg neurons, where depolarization enhanced the amplitude, duration and area under the curve (A.U.C.) of I-mGluR-evoked ER-mediated calcium signals when compared with basal conditions, this was not apparent in 3xTg-AD neurons. Whilst the amplitude of depolarization-enhanced I-mGluR-evoked ER-mediated calcium signals from both non-Tg F344 and TgF344-AD neurons was significantly enhanced relative to basal conditions, the A.U.C. and duration of responses were enhanced significantly upon depolarization in non-Tg F344, but not in TgF344-AD, neurons. Overall, the nature of basal I-mGluR-mediated calcium responses did not differ significantly between non-Tg F344 and TgF344-AD neurons. In summary, our results characterizing ER- and nSOCE-mediated calcium signaling in neurons demonstrate that ER Ca2+ dyshomeostasis is an early and potentially pathogenic event in familial AD.
Collapse
Affiliation(s)
- Aidan Kaar
- Department of Physiology, School of Medicine, University College Cork, Western Gateway Building, Cork, Ireland
| | - Megan P Weir
- Department of Physiology, School of Medicine, University College Cork, Western Gateway Building, Cork, Ireland
| | - Mark G Rae
- Department of Physiology, School of Medicine, University College Cork, Western Gateway Building, Cork, Ireland.
| |
Collapse
|
3
|
Love RWB. Aniracetam: An Evidence-Based Model for Preventing the Accumulation of Amyloid-β Plaques in Alzheimer's Disease. J Alzheimers Dis 2024; 98:1235-1241. [PMID: 38552113 DOI: 10.3233/jad-231247] [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] [Indexed: 04/20/2024]
Abstract
Alzheimer's disease is the leading cause of dementia in the world. It affects 6 million people in the United States and 50 million people worldwide. Alzheimer's disease is characterized by the accumulation of amyloid-β plaques (Aβ), an increase in tau protein neurofibrillary tangles, and a loss of synapses. Since the 1990s, removing and reducing Aβ has been the focus of Alzheimer's treatment and prevention research. The accumulation of Aβ can lead to oxidative stress, inflammation, neurotoxicity, and eventually apoptosis. These insults impair signaling systems in the brain, potentially leading to memory loss and cognitive decline. Aniracetam is a safe, effective, cognitive-enhancing drug that improves memory in both human and animal studies. Aniracetam may prevent the production and accumulation of Aβ by increasing α-secretase activity through two distinct pathways: 1) increasing brain derived neurotrophic factor expression and 2) positively modulating metabotropic glutamate receptors. This is the first paper to propose an evidence-based model for aniracetam reducing the accumulation and production of Aβ.
Collapse
Affiliation(s)
- Robert W B Love
- Research Department, Brain Fit For Life, LLC, Lewes, DE, USA
| |
Collapse
|
4
|
Gautam D, Naik UP, Naik MU, Yadav SK, Chaurasia RN, Dash D. Glutamate Receptor Dysregulation and Platelet Glutamate Dynamics in Alzheimer's and Parkinson's Diseases: Insights into Current Medications. Biomolecules 2023; 13:1609. [PMID: 38002291 PMCID: PMC10669830 DOI: 10.3390/biom13111609] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2023] [Revised: 10/31/2023] [Accepted: 11/01/2023] [Indexed: 11/26/2023] Open
Abstract
Two of the most prevalent neurodegenerative disorders (NDDs), Alzheimer's disease (AD) and Parkinson's disease (PD), present significant challenges to healthcare systems worldwide. While the etiologies of AD and PD differ, both diseases share commonalities in synaptic dysfunction, thereby focusing attention on the role of neurotransmitters. The possible functions that platelets may play in neurodegenerative illnesses including PD and AD are becoming more acknowledged. In AD, platelets have been investigated for their ability to generate amyloid-ß (Aß) peptides, contributing to the formation of neurotoxic plaques. Moreover, platelets are considered biomarkers for early AD diagnosis. In PD, platelets have been studied for their involvement in oxidative stress and mitochondrial dysfunction, which are key factors in the disease's pathogenesis. Emerging research shows that platelets, which release glutamate upon activation, also play a role in these disorders. Decreased glutamate uptake in platelets has been observed in Alzheimer's and Parkinson's patients, pointing to a systemic dysfunction in glutamate handling. This paper aims to elucidate the critical role that glutamate receptors play in the pathophysiology of both AD and PD. Utilizing data from clinical trials, animal models, and cellular studies, we reviewed how glutamate receptors dysfunction contributes to neurodegenerative (ND) processes such as excitotoxicity, synaptic loss, and cognitive impairment. The paper also reviews all current medications including glutamate receptor antagonists for AD and PD, highlighting their mode of action and limitations. A deeper understanding of glutamate receptor involvement including its systemic regulation by platelets could open new avenues for more effective treatments, potentially slowing disease progression and improving patient outcomes.
Collapse
Affiliation(s)
- Deepa Gautam
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
- The Cardeza Foundation for Hematologic Research, Center for Hemostasis, Thrombosis and Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (U.P.N.); (M.U.N.); (S.K.Y.)
| | - Ulhas P. Naik
- The Cardeza Foundation for Hematologic Research, Center for Hemostasis, Thrombosis and Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (U.P.N.); (M.U.N.); (S.K.Y.)
| | - Meghna U. Naik
- The Cardeza Foundation for Hematologic Research, Center for Hemostasis, Thrombosis and Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (U.P.N.); (M.U.N.); (S.K.Y.)
| | - Santosh K. Yadav
- The Cardeza Foundation for Hematologic Research, Center for Hemostasis, Thrombosis and Vascular Biology, Department of Medicine, Thomas Jefferson University, Philadelphia, PA 19107, USA; (U.P.N.); (M.U.N.); (S.K.Y.)
| | - Rameshwar Nath Chaurasia
- The Department of Neurology, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India;
| | - Debabrata Dash
- Center for Advanced Research on Platelet Signaling and Thrombosis Biology, Department of Biochemistry, Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| |
Collapse
|
5
|
Baracaldo-Santamaría D, Avendaño-Lopez SS, Ariza-Salamanca DF, Rodriguez-Giraldo M, Calderon-Ospina CA, González-Reyes RE, Nava-Mesa MO. Role of Calcium Modulation in the Pathophysiology and Treatment of Alzheimer's Disease. Int J Mol Sci 2023; 24:ijms24109067. [PMID: 37240413 DOI: 10.3390/ijms24109067] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2023] [Revised: 05/02/2023] [Accepted: 05/05/2023] [Indexed: 05/28/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disease and the most frequent cause of progressive dementia in senior adults. It is characterized by memory loss and cognitive impairment secondary to cholinergic dysfunction and N-methyl-D-aspartate (NMDA)-mediated neurotoxicity. Intracellular neurofibrillary tangles, extracellular plaques composed of amyloid-β (Aβ), and selective neurodegeneration are the anatomopathological hallmarks of this disease. The dysregulation of calcium may be present in all the stages of AD, and it is associated with other pathophysiological mechanisms, such as mitochondrial failure, oxidative stress, and chronic neuroinflammation. Although the cytosolic calcium alterations in AD are not completely elucidated, some calcium-permeable channels, transporters, pumps, and receptors have been shown to be involved at the neuronal and glial levels. In particular, the relationship between glutamatergic NMDA receptor (NMDAR) activity and amyloidosis has been widely documented. Other pathophysiological mechanisms involved in calcium dyshomeostasis include the activation of L-type voltage-dependent calcium channels, transient receptor potential channels, and ryanodine receptors, among many others. This review aims to update the calcium-dysregulation mechanisms in AD and discuss targets and molecules with therapeutic potential based on their modulation.
Collapse
Affiliation(s)
- Daniela Baracaldo-Santamaría
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Sara Sofia Avendaño-Lopez
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Daniel Felipe Ariza-Salamanca
- Medical and Health Sciences Education Research Group, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Mateo Rodriguez-Giraldo
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| | - Carlos A Calderon-Ospina
- Pharmacology Unit, Department of Biomedical Sciences, School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
- Grupo de Investigación en Ciencias Biomédicas Aplicadas (UR Biomed), School of Medicine and Health Sciences, Universidad del Rosario, Bogotá 111221, Colombia
| | - Rodrigo E González-Reyes
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| | - Mauricio O Nava-Mesa
- Grupo de Investigación en Neurociencias (NeURos), Centro de Neurociencias Neurovitae-UR, Instituto de Medicina Traslacional (IMT), Escuela de Medicina y Ciencias de la Salud, Universidad del Rosario, Bogotá 111221, Colombia
| |
Collapse
|
6
|
Holter KM, Pierce BE, Gould RW. Metabotropic glutamate receptor function and regulation of sleep-wake cycles. INTERNATIONAL REVIEW OF NEUROBIOLOGY 2023; 168:93-175. [PMID: 36868636 PMCID: PMC10973983 DOI: 10.1016/bs.irn.2022.11.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
Metabotropic glutamate (mGlu) receptors are the most abundant family of G-protein coupled receptors and are widely expressed throughout the central nervous system (CNS). Alterations in glutamate homeostasis, including dysregulations in mGlu receptor function, have been indicated as key contributors to multiple CNS disorders. Fluctuations in mGlu receptor expression and function also occur across diurnal sleep-wake cycles. Sleep disturbances including insomnia are frequently comorbid with neuropsychiatric, neurodevelopmental, and neurodegenerative conditions. These often precede behavioral symptoms and/or correlate with symptom severity and relapse. Chronic sleep disturbances may also be a consequence of primary symptom progression and can exacerbate neurodegeneration in disorders including Alzheimer's disease (AD). Thus, there is a bidirectional relationship between sleep disturbances and CNS disorders; disrupted sleep may serve as both a cause and a consequence of the disorder. Importantly, comorbid sleep disturbances are rarely a direct target of primary pharmacological treatments for neuropsychiatric disorders even though improving sleep can positively impact other symptom clusters. This chapter details known roles of mGlu receptor subtypes in both sleep-wake regulation and CNS disorders focusing on schizophrenia, major depressive disorder, post-traumatic stress disorder, AD, and substance use disorder (cocaine and opioid). In this chapter, preclinical electrophysiological, genetic, and pharmacological studies are described, and, when possible, human genetic, imaging, and post-mortem studies are also discussed. In addition to reviewing the important relationships between sleep, mGlu receptors, and CNS disorders, this chapter highlights the development of selective mGlu receptor ligands that hold promise for improving both primary symptoms and sleep disturbances.
Collapse
Affiliation(s)
- Kimberly M Holter
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Bethany E Pierce
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States
| | - Robert W Gould
- Department of Physiology and Pharmacology, Wake Forest University School of Medicine, Winston-Salem, NC, United States.
| |
Collapse
|
7
|
Khotimchenko YS, Silachev DN, Katanaev VL. Marine Natural Products from the Russian Pacific as Sources of Drugs for Neurodegenerative Diseases. Mar Drugs 2022; 20:708. [PMID: 36421986 PMCID: PMC9697637 DOI: 10.3390/md20110708] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2022] [Revised: 11/07/2022] [Accepted: 11/09/2022] [Indexed: 09/05/2023] Open
Abstract
Neurodegenerative diseases are growing to become one of humanity's biggest health problems, given the number of individuals affected by them. They cause enough mortalities and severe economic impact to rival cancers and infections. With the current diversity of pathophysiological mechanisms involved in neurodegenerative diseases, on the one hand, and scarcity of efficient prevention and treatment strategies, on the other, all possible sources for novel drug discovery must be employed. Marine pharmacology represents a relatively uncharted territory to seek promising compounds, despite the enormous chemodiversity it offers. The current work discusses one vast marine region-the Northwestern or Russian Pacific-as the treasure chest for marine-based drug discovery targeting neurodegenerative diseases. We overview the natural products of neurological properties already discovered from its waters and survey the existing molecular and cellular targets for pharmacological modulation of the disease. We further provide a general assessment of the drug discovery potential of the Russian Pacific in case of its systematic development to tackle neurodegenerative diseases.
Collapse
Affiliation(s)
- Yuri S. Khotimchenko
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 8 ul. Sukhanova, 690950 Vladivostok, Russia
- A.V. Zhirmunsky National Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, 690950 Vladivostok, Russia
| | - Denis N. Silachev
- Department of Functional Biochemistry of Biopolymers, A.N. Belozersky Research Institute of Physico-Chemical Biology, Moscow State University, 119992 Moscow, Russia
| | - Vladimir L. Katanaev
- Institute of Life Sciences and Biomedicine, Far Eastern Federal University, 8 ul. Sukhanova, 690950 Vladivostok, Russia
- Department of Cell Physiology and Metabolism, Translational Research Centre in Oncohaematology, Faculty of Medicine, University of Geneva, Rue Michel-Servet 1, 1211 Geneva, Switzerland
| |
Collapse
|
8
|
Li SH, Abd-Elrahman KS, Ferguson SS. Targeting mGluR2/3 for treatment of neurodegenerative and neuropsychiatric diseases. Pharmacol Ther 2022; 239:108275. [DOI: 10.1016/j.pharmthera.2022.108275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2022] [Revised: 08/23/2022] [Accepted: 08/23/2022] [Indexed: 10/15/2022]
|
9
|
Fan S, Li L, Liu L, Li H, Xian X, Li W. Ceftriaxone Suppresses Group II Metabotropic Glutamate Receptor Expression Contributing to Reversal of Recognition Memory Deficits of Amyloid Precursor Protein/Presenilin 1 AD Mice. Front Neurosci 2022; 16:905403. [PMID: 35860293 PMCID: PMC9289516 DOI: 10.3389/fnins.2022.905403] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/27/2022] [Accepted: 06/01/2022] [Indexed: 11/13/2022] Open
Abstract
Group II metabotropic glutamate receptors (Group II mGluRs) are the peri-synaptic receptor of glutamatergic neurons and negatively regulate glutamate release from presynaptic neurons. Glutamate in the synaptic cleft is mainly taken into astrocytes by glutamate transporter-1 (GLT-1), which is primarily expressed in astrocytes. Increasing evidence showed that inhibiting or suppressing the activation of Group II mGluRs would contribute to the improvement of learning and memory deficits in Alzheimer’s disease (AD) animal models. Ceftriaxone (Cef) has been reported to alleviate the spatial memory deficits in AD model mice by improving GLT-1-related clearance and metabolism of glutamate. Therefore, the present study further investigates the improving effect of Cef on recognition memory deficits and the involvement of Group II mGluRs in the process using the APP/PS1 AD mouse model. Novel object recognition tests showed that the Cef treatment significantly improved the recognition memory deficits of the AD mice. The Western blot and immunohistochemistry analysis showed that the Cef treatment significantly suppressed the upregulation of Group II mGluRs expression in APP/PS1 AD mice. The above suppression effect of Cef was blocked by dihydrokainic acid, an inhibitor of GLT-1 uptake activity. Furthermore, the Cef treatment significantly restored the downregulation in the downstream molecules of Group II mGluRs activation, including the expression of PKA and phosphorylated SNAP-25 in the APP/PS1 AD mice. The Cef treatment had no effect on the content of Aβ40 and Aβ42 in the hippocampus of APP/PS1 AD mice. The above results suggested that the suppression of Group II mGluRs contributed to the Cef-induced reversal of the recognition memory deficits in APP/PS1 AD mice.
Collapse
Affiliation(s)
- ShuJuan Fan
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - Li Li
- Department of Central Laboratory, The Second Hospital of Hebei Medical University, Shijiazhuang, China
| | - LiRong Liu
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - He Li
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
| | - XiaoHui Xian
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
- *Correspondence: XiaoHui Xian,
| | - WenBin Li
- Department of Pathophysiology, Neuroscience Research Center, Hebei Medical University, Shijiazhuang, China
- WenBin Li,
| |
Collapse
|
10
|
Jia Y, Wang X, Chen Y, Qiu W, Ge W, Ma C. Proteomic and Transcriptomic Analyses Reveal Pathological Changes in the Entorhinal Cortex Region that Correlate Well with Dysregulation of Ion Transport in Patients with Alzheimer's Disease. Mol Neurobiol 2021; 58:4007-4027. [PMID: 33904022 DOI: 10.1007/s12035-021-02356-3] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/04/2020] [Accepted: 03/10/2021] [Indexed: 01/17/2023]
Abstract
Alzheimer's disease (AD) is the most common neurodegenerative disorder. The earliest neuropathology of AD appears in entorhinal cortex (EC) regions. Therapeutic strategies and preventive measures to protect against entorhinal degeneration would be of substantial value in the early stages of AD. In this study, transcriptome based on the Illumina RNA-seq and proteome based on TMT-labelling were performed for RNA and protein profiling on AD EC samples and non-AD control EC samples. Immunohistochemistry was used to validate proteins expressions. After integrated analysis, 57 genes were detected both in transcriptome and proteome data, including 51 in similar altering trends (7 upregulated, 44 downregulated) and 6 in inverse trends when compared AD vs. control. The top 6 genes (GABRG2, CACNG3, CACNB4, GABRB2, GRIK2, and SLC17A6) within the 51 genes were selected and related to "ion transport". Correlation analysis demonstrated negative relationship of protein expression level with the neuropathologic changes. In conclusion, the integrate transcriptome and proteome analysis provided evidence for dysregulation of ion transport across brain regions in AD, which might be a critical signaling pathway that initiates pathology. This study might provide new insight into the earliest changes occurring in the EC of AD and novel targets for AD prevention and treatment.
Collapse
Affiliation(s)
- Yangjie Jia
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, National Human Brain Bank for Development and Function, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China
| | - Xia Wang
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China
| | - Yanyu Chen
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China
| | - Wenying Qiu
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, National Human Brain Bank for Development and Function, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China
| | - Wei Ge
- State Key Laboratory of Medical Molecular Biology and Department of Immunology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China.
| | - Chao Ma
- Department of Human Anatomy, Histology and Embryology, Neuroscience Center, National Human Brain Bank for Development and Function, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences, Peking Union Medical College, No. 5 Dongdansantiao, Dongcheng District, Beijing, 100005, China.
| |
Collapse
|
11
|
Gasiorowska A, Wydrych M, Drapich P, Zadrozny M, Steczkowska M, Niewiadomski W, Niewiadomska G. The Biology and Pathobiology of Glutamatergic, Cholinergic, and Dopaminergic Signaling in the Aging Brain. Front Aging Neurosci 2021; 13:654931. [PMID: 34326765 PMCID: PMC8315271 DOI: 10.3389/fnagi.2021.654931] [Citation(s) in RCA: 68] [Impact Index Per Article: 22.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2021] [Accepted: 06/14/2021] [Indexed: 12/12/2022] Open
Abstract
The elderly population is growing worldwide, with important health and socioeconomic implications. Clinical and experimental studies on aging have uncovered numerous changes in the brain, such as decreased neurogenesis, increased synaptic defects, greater metabolic stress, and enhanced inflammation. These changes are associated with cognitive decline and neurobehavioral deficits. Although aging is not a disease, it is a significant risk factor for functional worsening, affective impairment, disease exaggeration, dementia, and general disease susceptibility. Conversely, life events related to mental stress and trauma can also lead to accelerated age-associated disorders and dementia. Here, we review human studies and studies on mice and rats, such as those modeling human neurodegenerative diseases, that have helped elucidate (1) the dynamics and mechanisms underlying the biological and pathological aging of the main projecting systems in the brain (glutamatergic, cholinergic, and dopaminergic) and (2) the effect of defective glutamatergic, cholinergic, and dopaminergic projection on disabilities associated with aging and neurodegenerative disorders, such as Alzheimer's and Parkinson's diseases. Detailed knowledge of the mechanisms of age-related diseases can be an important element in the development of effective ways of treatment. In this context, we briefly analyze which adverse changes associated with neurodegenerative diseases in the cholinergic, glutaminergic and dopaminergic systems could be targeted by therapeutic strategies developed as a result of our better understanding of these damaging mechanisms.
Collapse
Affiliation(s)
- Anna Gasiorowska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Malgorzata Wydrych
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Patrycja Drapich
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Maciej Zadrozny
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Marta Steczkowska
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Wiktor Niewiadomski
- Mossakowski Medical Research Institute, Polish Academy of Sciences, Warsaw, Poland
| | - Grazyna Niewiadomska
- Nencki Institute of Experimental Biology, Polish Academy of Sciences, Warsaw, Poland
| |
Collapse
|
12
|
Qunies AM, Emmitte KA. Negative allosteric modulators of group II metabotropic glutamate receptors: A patent review (2015 - present). Expert Opin Ther Pat 2021; 31:687-708. [PMID: 33719801 DOI: 10.1080/13543776.2021.1903431] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
INTRODUCTION Group II metabotropic glutamate (mGlu) receptors have emerged as an attractive potential target for the development of novel CNS therapeutics in areas such as Alzheimer's disease (AD), anxiety, cognitive disorders, depression, and others. Several small molecules that act as negative allosteric modulators (NAMs) on these receptors have demonstrated efficacy and/or target engagement in animal models, and one molecule (decoglurant) has been advanced into clinical trials. AREAS COVERED This review summarizes patent applications published between January 2015 and November 2020. It is divided into three sections: (1) small molecule nonselective mGlu2/3 NAMs, (2) small molecule selective mGlu2 NAMs, and (3) small molecule selective mGlu3 NAMs. EXPERT OPINION Much progress has been made in the discovery of novel small molecule mGlu2 NAMs. Still, chemical diversity remains somewhat limited and room for expansion remains. Progress with mGlu3 NAMs has been more limited; however, some promising molecules have been disclosed. The process of elucidating the precise role of each receptor in the diseases associated with group II receptors has begun. Continued studies in animals with selective NAMs for both receptors will be critical in the coming years to inform researchers on the right compound profile and patient population for clinical development.
Collapse
Affiliation(s)
- Alshaima'a M Qunies
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, USA.,Graduate School of Biomedical Sciences, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Kyle A Emmitte
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, Fort Worth, TX, USA
| |
Collapse
|
13
|
Shabani S. A mechanistic view on the neurotoxic effects of air pollution on central nervous system: risk for autism and neurodegenerative diseases. ENVIRONMENTAL SCIENCE AND POLLUTION RESEARCH INTERNATIONAL 2021; 28:6349-6373. [PMID: 33398761 DOI: 10.1007/s11356-020-11620-3] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/26/2019] [Accepted: 11/09/2020] [Indexed: 06/12/2023]
Abstract
Many reports have shown a strong association between exposure to neurotoxic air pollutants like heavy metal and particulate matter (PM) as an active participant and neurological disorders. While the effects of these toxic pollutants on cardiopulmonary morbidity have principally been studied, growing evidence has shown that exposure to polluted air is associated with memory impairment, communication deficits, and anxiety/depression among all ages. So, these toxic pollutants in the environment increase the risk of neurodegenerative disease, ischemia, and autism spectrum disorders (ASD). The precise mechanisms in which air pollutants lead to communicative inability, social inability, and declined cognition have remained unknown. Various animal model studies show that amyloid precursor protein (APP), processing, oxidant/antioxidant balance, and inflammation pathways change following the exposure to constituents of polluted air. In the present review study, we collect the probable molecular mechanisms of deleterious CNS effects in response to various air pollutants.
Collapse
Affiliation(s)
- Sahreh Shabani
- Medical Plants Research Center, Basic Health Sciences Institute, Shahrekord University of Medical Sciences, Shahrekord, Iran.
| |
Collapse
|
14
|
Bukke VN, Archana M, Villani R, Romano AD, Wawrzyniak A, Balawender K, Orkisz S, Beggiato S, Serviddio G, Cassano T. The Dual Role of Glutamatergic Neurotransmission in Alzheimer's Disease: From Pathophysiology to Pharmacotherapy. Int J Mol Sci 2020; 21:ijms21207452. [PMID: 33050345 PMCID: PMC7589203 DOI: 10.3390/ijms21207452] [Citation(s) in RCA: 63] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2020] [Revised: 09/28/2020] [Accepted: 10/05/2020] [Indexed: 12/19/2022] Open
Abstract
Alzheimer's disease (AD) is an age-related dementia and neurodegenerative disorder, characterized by Aβ and tau protein deposition impairing learning, memory and suppressing synaptic plasticity of neurons. Increasing evidence suggests that there is a link between the glucose and glutamate alterations with age that down-regulates glucose utilization reducing glutamate levels in AD patients. Deviations in brain energy metabolism reinforce the development of AD by hampering glutamate levels in the brain. Glutamate is a nonessential amino acid and the major excitatory neurotransmitter synthesized from glucose. Alterations in cerebral glucose and glutamate levels precede the deposition of Aβ plaques. In the brain, over 40% of neuronal synapses are glutamatergic and disturbances in glutamatergic function have been implicated in pathophysiology of AD. Nevertheless, targeting the glutamatergic system seems to be a promising strategy to develop novel, improved therapeutics for AD. Here, we review data supporting the involvement of the glutamatergic system in AD pathophysiology as well as the efficacy of glutamatergic agents in this neurodegenerative disorder. We also discuss exciting new prospects for the development of improved therapeutics for this devastating disorder.
Collapse
Affiliation(s)
- Vidyasagar Naik Bukke
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
| | - Moola Archana
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.A.); (R.V.); (A.D.R.); (G.S.)
| | - Rosanna Villani
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.A.); (R.V.); (A.D.R.); (G.S.)
| | - Antonino Davide Romano
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.A.); (R.V.); (A.D.R.); (G.S.)
| | - Agata Wawrzyniak
- Morphological Science Department of Human Anatomy, Medical Faculty University of Rzeszów, 35-310 Rzeszów, Poland; (A.W.); (K.B.); (S.O.)
| | - Krzysztof Balawender
- Morphological Science Department of Human Anatomy, Medical Faculty University of Rzeszów, 35-310 Rzeszów, Poland; (A.W.); (K.B.); (S.O.)
| | - Stanislaw Orkisz
- Morphological Science Department of Human Anatomy, Medical Faculty University of Rzeszów, 35-310 Rzeszów, Poland; (A.W.); (K.B.); (S.O.)
| | - Sarah Beggiato
- Department of Medical, Oral and Biotechnological Sciences, University of Chieti-Pescara, 66100 Chieti, Italy;
| | - Gaetano Serviddio
- Department of Medical and Surgical Sciences, University of Foggia, 71122 Foggia, Italy; (M.A.); (R.V.); (A.D.R.); (G.S.)
| | - Tommaso Cassano
- Department of Clinical and Experimental Medicine, University of Foggia, 71122 Foggia, Italy;
- Correspondence:
| |
Collapse
|
15
|
Yuan G, Qu X, Zheng B, Neelamegam R, Afshar S, Iyengar S, Pan C, Wang J, Kang HJ, Ondrechen MJ, Poutiainen P, El Fakhri G, Zhang Z, Brownell AL. Design, Synthesis, and Characterization of Benzimidazole Derivatives as Positron Emission Tomography Imaging Ligands for Metabotropic Glutamate Receptor 2. J Med Chem 2020; 63:12060-12072. [PMID: 32981322 DOI: 10.1021/acs.jmedchem.0c01394] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Three benzimidazole derivatives (13-15) have been synthetized as potential positron emission tomography (PET) imaging ligands for mGluR2 in the brain. Of these compounds, 13 exhibits potent binding affinity (IC50 = 7.6 ± 0.9 nM), positive allosteric modulator (PAM) activity (EC50 = 51.2 nM), and excellent selectivity against other mGluR subtypes (>100-fold). [11C]13 was synthesized via O-[11C]methylation of its phenol precursor 25 with [11C]methyl iodide. The achieved radiochemical yield was 20 ± 2% (n = 10, decay-corrected) based on [11C]CO2 with a radiochemical purity of >98% and molar activity of 98 ± 30 GBq/μmol EOS. Ex vivo biodistribution studies revealed reversible accumulation of [11C]13 and hepatobiliary and urinary excretions. PET imaging studies in rats demonstrated that [11C]13 accumulated in the mGluR2-rich brain regions. Pre-administration of mGluR2-selective PAM, 17 reduced the brain uptake of [11C]13, indicating a selective binding. Therefore, [11C]13 is a potential PET imaging ligand for mGluR2 in different central nervous system-related conditions.
Collapse
Affiliation(s)
- Gengyang Yuan
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Xiying Qu
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Baohui Zheng
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Ramesh Neelamegam
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Sepideh Afshar
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Suhasini Iyengar
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Chuzhi Pan
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Junfeng Wang
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Hye Jin Kang
- Department of Pharmacology, University of North Carolina Chapel Hill School of Medicine, Chapel Hill, North Carolina 27514, United States
| | - Mary Jo Ondrechen
- Department of Chemistry and Chemical Biology, Northeastern University, 360 Huntington Avenue, Boston, Massachusetts 02115, United States
| | - Pekka Poutiainen
- Department of Clinical Physiology and Nuclear Medicine, Kuopio University Hospital, Kuopio 70210, Finland
| | - Georges El Fakhri
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| | - Zhaoda Zhang
- Athinoula A. Martinos Center for Biomedical Imaging, Massachusetts General Hospital and Harvard Medical School, 149 Thirteenth Street, Suite 2301, Charlestown, Massachusetts 02129, United States
| | - Anna-Liisa Brownell
- Gordon Center for Medical Imaging, Massachusetts General Hospital and Harvard Medical School, 3rd Avenue, Charlestown, Massachusetts 02129, United States
| |
Collapse
|
16
|
Hascup KN, Britz J, Findley CA, Tischkau S, Hascup ER. LY379268 Does Not Have Long-Term Procognitive Effects nor Attenuate Glutamatergic Signaling in AβPP/PS1 Mice. J Alzheimers Dis 2020; 68:1193-1209. [PMID: 30909243 DOI: 10.3233/jad-181231] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Chronically elevated basal glutamate levels are hypothesized to attenuate detection of physiological signals thereby inhibiting memory formation and retrieval, while inducing excitotoxicity-mediated neurodegeneration observed in Alzheimer's disease (AD). However, current medication targeting the glutamatergic system, such as memantine, shows limited efficacy and is unable to decelerate disease progression, possibly because it modulates postsynaptic N-methyl-D-aspartate receptors rather than glutamate release or clearance. To determine if decreasing presynaptic glutamate release leads to long-term procognitive effects, we treated AβPP/PS1 mice with LY379268 (3.0 mg/kg; i.p.), a metabotropic glutamate receptor (mGluR)2/3 agonist from 2-6 months of age when elevated glutamate levels are first observed but cognition is unaffected. C57BL/6J genetic background control mice and another cohort of AβPP/PS1 mice received normal saline (i.p.) as vehicle controls. After 6 months off treatment, mice receiving LY379268 did not show long-term improvement as assessed by the Morris water maze (MWM) spatial learning and memory paradigm. Following MWM, mice were isoflurane anesthetized and a glutamate selective microelectrode was used to measure in vivo basal and stimulus-evoked glutamate release and clearance independently from the dentate, CA3, and CA1 hippocampal subregions. Immunohistochemistry was used to measure hippocampal astrogliosis and plaque pathology. Similar to previous studies, we observed elevated basal glutamate, stimulus evoked glutamate release, and astrogliosis in AβPP/PS1 vehicle mice versus C57BL/6J mice. Treatment with LY379268 did not attenuate these responses nor diminish plaque pathology. The current study builds upon previous research demonstrating hyperglutamatergic hippocampal signaling in AβPP/PS1 mice; however, long-term therapeutic efficacy of LY379268 in AβPP/PS1 was not observed.
Collapse
Affiliation(s)
- Kevin N Hascup
- Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neurosciences Institute, Springfield, IL, USA.,Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Jesse Britz
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Caleigh A Findley
- Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neurosciences Institute, Springfield, IL, USA.,Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Shelley Tischkau
- Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| | - Erin R Hascup
- Department of Neurology, Center for Alzheimer's Disease and Related Disorders, Neurosciences Institute, Springfield, IL, USA.,Department of Pharmacology, Southern Illinois University School of Medicine, Springfield, IL, USA
| |
Collapse
|
17
|
Srivastava A, Das B, Yao AY, Yan R. Metabotropic Glutamate Receptors in Alzheimer's Disease Synaptic Dysfunction: Therapeutic Opportunities and Hope for the Future. J Alzheimers Dis 2020; 78:1345-1361. [PMID: 33325389 PMCID: PMC8439550 DOI: 10.3233/jad-201146] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Alzheimer's disease (AD) is a neurodegenerative disorder characterized by the presence of neuritic plaques and neurofibrillary tangles. The impaired synaptic plasticity and dendritic loss at the synaptic level is an early event associated with the AD pathogenesis. The abnormal accumulation of soluble oligomeric amyloid-β (Aβ), the major toxic component in amyloid plaques, is viewed to trigger synaptic dysfunctions through binding to several presynaptic and postsynaptic partners and thus to disrupt synaptic transmission. Over time, the abnormalities in neural transmission will result in cognitive deficits, which are commonly manifested as memory loss in AD patients. Synaptic plasticity is regulated through glutamate transmission, which is mediated by various glutamate receptors. Here we review recent progresses in the study of metabotropic glutamate receptors (mGluRs) in AD cognition. We will discuss the role of mGluRs in synaptic plasticity and their modulation as a possible strategy for AD cognitive improvement.
Collapse
Affiliation(s)
- Akriti Srivastava
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Brati Das
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Annie Y. Yao
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| | - Riqiang Yan
- Department of Neuroscience, University of Connecticut Health, Farmington, CT, USA
| |
Collapse
|
18
|
Maesako M, Zoltowska KM, Berezovska O. Synapsin 1 promotes Aβ generation via BACE1 modulation. PLoS One 2019; 14:e0226368. [PMID: 31830091 PMCID: PMC6907790 DOI: 10.1371/journal.pone.0226368] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2019] [Accepted: 11/25/2019] [Indexed: 12/26/2022] Open
Abstract
It has been revealed that β-amyloid (Aβ) is generated and released from the presynaptic terminals in activity-dependent manner. However, molecules modulating the presynaptic Aβ generation remain elusive. Here we test the hypothesis that Synapsin 1 (Syn1) may acts as a modulator of the Aβ production. Using biochemical and Förster resonance energy transfer (FRET)-based imaging approaches we have found that Syn1 knock down decreases, whereas (over)expression of Syn1 in cells increases the Aβ levels. Mechanistically, Syn1 does not seem to affect the activity of Presenilin 1 (PS1)/γ-secretase, PS1 conformation, or the proximity between PS1 and amyloid precursor protein (APP). However, we found that Syn1 is involved in up-regulation of the β-site APP cleaving enzyme 1 (BACE1)/β-secretase activity and increases the APP/BACE1 interaction. Therefore, we conclude that Syn1 may promote Aβ production via the modulation of BACE1.
Collapse
Affiliation(s)
- Masato Maesako
- MassGeneral Institute for Neurodegenerative Disease, Alzheimer’s Disease Research Unit, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States of America
| | - Katarzyna M. Zoltowska
- MassGeneral Institute for Neurodegenerative Disease, Alzheimer’s Disease Research Unit, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States of America
| | - Oksana Berezovska
- MassGeneral Institute for Neurodegenerative Disease, Alzheimer’s Disease Research Unit, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, United States of America
- * E-mail:
| |
Collapse
|
19
|
Jin LE, Wang M, Galvin VC, Lightbourne TC, Conn PJ, Arnsten AFT, Paspalas CD. mGluR2 versus mGluR3 Metabotropic Glutamate Receptors in Primate Dorsolateral Prefrontal Cortex: Postsynaptic mGluR3 Strengthen Working Memory Networks. Cereb Cortex 2019; 28:974-987. [PMID: 28108498 DOI: 10.1093/cercor/bhx005] [Citation(s) in RCA: 77] [Impact Index Per Article: 15.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2016] [Indexed: 01/06/2023] Open
Abstract
The newly evolved circuits in layer III of primate dorsolateral prefrontal cortex (dlPFC) generate the neural representations that subserve working memory. These circuits are weakened by increased cAMP-K+ channel signaling, and are a focus of pathology in schizophrenia, aging, and Alzheimer's disease. Cognitive deficits in these disorders are increasingly associated with insults to mGluR3 metabotropic glutamate receptors, while reductions in mGluR2 appear protective. This has been perplexing, as mGluR3 has been considered glial receptors, and mGluR2 and mGluR3 have been thought to have similar functions, reducing glutamate transmission. We have discovered that, in addition to their astrocytic expression, mGluR3 is concentrated postsynaptically in spine synapses of layer III dlPFC, positioned to strengthen connectivity by inhibiting postsynaptic cAMP-K+ channel actions. In contrast, mGluR2 is principally presynaptic as expected, with only a minor postsynaptic component. Functionally, increase in the endogenous mGluR3 agonist, N-acetylaspartylglutamate, markedly enhanced dlPFC Delay cell firing during a working memory task via inhibition of cAMP signaling, while the mGluR2 positive allosteric modulator, BINA, produced an inverted-U dose-response on dlPFC Delay cell firing and working memory performance. These data illuminate why insults to mGluR3 would erode cognitive abilities, and support mGluR3 as a novel therapeutic target for higher cognitive disorders.
Collapse
Affiliation(s)
- Lu E Jin
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Min Wang
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Veronica C Galvin
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Taber C Lightbourne
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | - Peter Jeffrey Conn
- Department of Pharmacology and Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt UniversityMedical Center, Nashville, TN 37232-0697, USA
| | - Amy F T Arnsten
- Department of Neuroscience, Yale University School of Medicine, New Haven, CT 06510, USA
| | | |
Collapse
|
20
|
Abstract
Abnormalities of glutamatergic transmission are implicated in neuropsychiatric disorders. Among the glutamate receptors, metabotropic (mGlu) 2/3 receptors have recently gained much attention as molecular targets for the treatment of several neuropsychiatric disorders including depression and anxiety. Both orthosteric and allosteric antagonists of mGlu2/3 receptors have been synthesized, and their therapeutic potential has been examined. These research activities have demonstrated the promise of mGlu2/3 receptor antagonists as potential treatment agents for the above-mentioned neuropsychiatric disorders. In particular, it has been considered that the antidepressant effects of mGlu2/3 receptor antagonists are worthy of pursuing, since the antidepressant profiles as well as synaptic/neural mechanisms involved in the actions of mGlu2/3 receptor antagonists are similar to those of ketamine, which has been demonstrated to show potent, rapid and sustained efficacy in patients with depression, even those resistant to the conventionally prescribed antidepressants. In this chapter, the general pharmacology of mGlu2/3 receptor antagonists and their therapeutic potential are reviewed. In particular, I focus on the usefulness of mGlu2/3 receptor antagonists as novel antidepressants, in comparison with ketamine.
Collapse
|
21
|
Ourdev D, Schmaus A, Kar S. Kainate Receptor Activation Enhances Amyloidogenic Processing of APP in Astrocytes. Mol Neurobiol 2018; 56:5095-5110. [PMID: 30484111 DOI: 10.1007/s12035-018-1427-8] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2018] [Accepted: 11/13/2018] [Indexed: 12/21/2022]
Abstract
Kainic acid (KA) is an analogue of the excitatory neurotransmitter glutamate that, when injected systemically into adult rats, can trigger seizures and progressive neuronal loss in a manner that mirrors the neuropathology of human mesial temporal lobe epilepsy. However, biomolecular mechanisms responsible for the neuronal loss that occurs as a consequence of this treatment remains elusive. We have recently reported that toxicity induced by KA can partly be mediated by astrocyte-derived amyloid β (Aβ) peptides, which are critical in the development of Alzheimer's disease (AD). Nonetheless, little is known how KA can influence amyloid precursor protein (APP) levels and processing in astrocytes. Thus, in the present study using human U-373 astrocytoma and rat primary astrocytes, we evaluated the role of KA on APP metabolism. Our results revealed that KA treatment increased the levels of APP and its cleaved products (α-/β-CTFs) in cultured U-373 astrocytoma and primary astrocytes, without altering the cell viability. The cellular and secretory levels of Aβ1-40/Aβ1-42 were markedly increased in KA-treated astrocytes. We also demonstrated that the steady-state levels of APP-secretases were not altered but the activity of γ-secretase is enhanced in KA-treated U-373 astrocytoma. Furthermore, using selective receptor antagonists, we showed that the effects of KA is mediated by activation of kainate receptors and not NMDA or AMPA receptors. These results suggest that KA can enhance amyloidogenic processing of APP by activating its own receptor leading to increased production/secretion of Aβ-related peptides from activated astrocytes which may contribute to the pathogenesis of temporal lobe epilepsy.
Collapse
Affiliation(s)
- D Ourdev
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada.,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada
| | - A Schmaus
- Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada.,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada
| | - Satyabrata Kar
- Department of Psychiatry, University of Alberta, Edmonton, AB, Canada. .,Centre for Prions and Protein Folding Diseases, University of Alberta, Edmonton, AB, Canada. .,Neuroscience and Mental Health Institute, University of Alberta, Edmonton, AB, Canada. .,Department of Medicine, University of Alberta, Edmonton, AB, Canada.
| |
Collapse
|
22
|
Hettinger JC, Lee H, Bu G, Holtzman DM, Cirrito JR. AMPA-ergic regulation of amyloid-β levels in an Alzheimer's disease mouse model. Mol Neurodegener 2018; 13:22. [PMID: 29764453 PMCID: PMC5952376 DOI: 10.1186/s13024-018-0256-6] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/14/2017] [Accepted: 05/02/2018] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Extracellular aggregation of the amyloid-β (Aβ) peptide into toxic multimers is a key event in Alzheimer's disease (AD) pathogenesis. Aβ aggregation is concentration-dependent, with higher concentrations of Aβ much more likely to form toxic species. The processes that regulate extracellular levels of Aβ therefore stand to directly affect AD pathology onset. Studies from our lab and others have demonstrated that synaptic activity is a critical regulator of Aβ production through both presynaptic and postsynaptic mechanisms. AMPA receptors (AMPA-Rs), as the most abundant ionotropic glutamate receptors, have the potential to greatly impact Aβ levels. METHODS In order to study the role of AMPA-Rs in Aβ regulation, we used in vivo microdialysis in an APP/PS1 mouse model to simultaneously deliver AMPA and other treatments while collecting Aβ from the interstitial fluid (ISF). Changes in Aβ production and clearance along with inflammation were assessed using biochemical approaches. IL-6 deficient mice were utilized to test the role of IL-6 signaling in AMPA-R-mediated regulation of Aβ levels. RESULTS We found that AMPA-R activation decreases in ISF Aβ levels in a dose-dependent manner. Moreover, the effect of AMPA treatment involves three distinct pathways. Steady-state activity of AMPA-Rs normally promotes higher ISF Aβ. Evoked AMPA-R activity, however, decreases Aβ levels by both stimulating glutamatergic transmission and activating downstream NMDA receptor (NMDA-R) signaling and, with extended AMPA treatment, acting independently of NMDA-Rs. Surprisingly, we found this latter, direct AMPA pathway of Aβ regulation increases Aβ clearance, while Aβ production appears to be largely unaffected. Furthermore, the AMPA-dependent decrease is not observed in IL-6 deficient mice, indicating a role for IL-6 signaling in AMPA-R-mediated Aβ clearance. CONCLUSION Though basal levels of AMPA-R activity promote higher levels of ISF Aβ, evoked AMPA-R signaling decreases Aβ through both NMDA-R-dependent and -independent pathways. We find that evoked AMPA-R signaling increases clearance of extracellular Aβ, at least in part through enhanced IL-6 signaling. These data emphasize that Aβ regulation by synaptic activity involves a number of independent pathways that together determine extracellular Aβ levels. Understanding how these pathways maintain Aβ levels prior to AD pathology may provide insights into disease pathogenesis.
Collapse
Affiliation(s)
- Jane C Hettinger
- Department of Neurology, Knight Alzheimer's Disease Research Center, Hope Center for Neurological Disorders, Washington University School of Medicine, Campus Box 8111, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Hyo Lee
- Department of Neurology, Knight Alzheimer's Disease Research Center, Hope Center for Neurological Disorders, Washington University School of Medicine, Campus Box 8111, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - Guojun Bu
- Department of Neuroscience, Mayo Clinic, Jacksonville, FL, 32224, USA
| | - David M Holtzman
- Department of Neurology, Knight Alzheimer's Disease Research Center, Hope Center for Neurological Disorders, Washington University School of Medicine, Campus Box 8111, 660 South Euclid Avenue, St. Louis, MO, 63110, USA
| | - John R Cirrito
- Department of Neurology, Knight Alzheimer's Disease Research Center, Hope Center for Neurological Disorders, Washington University School of Medicine, Campus Box 8111, 660 South Euclid Avenue, St. Louis, MO, 63110, USA.
| |
Collapse
|
23
|
Hoshi A, Tsunoda A, Yamamoto T, Tada M, Kakita A, Ugawa Y. Altered expression of glutamate transporter-1 and water channel protein aquaporin-4 in human temporal cortex with Alzheimer's disease. Neuropathol Appl Neurobiol 2018; 44:628-638. [PMID: 29405337 DOI: 10.1111/nan.12475] [Citation(s) in RCA: 45] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2017] [Accepted: 01/11/2018] [Indexed: 12/13/2022]
Abstract
AIMS Glutamate neurotoxicity plays an important role in the pathogenesis of various neurodegenerative disorders. Many studies have demonstrated that glutamate transporter-1 (GLT-1), the dominant astrocytic glutamate transporter, is significantly reduced in the cerebral cortex of patients with Alzheimer's disease (AD), suggesting that glutamate-mediated excitotoxicity might contribute to the pathogenesis of AD. In a previous study, we have demonstrated marked alterations in the expression of the astrocytic water channel protein aquaporin-4 (AQP4) in relation to amyloid β deposition in human AD brains. As a functional complex, GLT-1 and AQP4 in astrocytes may play a neuroprotective role in the progression of AD pathology. However, few studies have examined the correlation between the expression of GLT-1 and that of AQP4 in human AD brain. METHODS Here, using immunohistochemistry with antibodies against GLT-1 and AQP4, we studied the expression levels and distribution patterns of GLT-1 in areas showing various patterns of AQP4 expression in autopsied temporal lobes from eight patients with AD and five controls without neurological disorders. RESULTS GLT-1 staining in the control group was present throughout the neocortex as uniform neuropil staining with co-localized AQP4. The AD group showed a significant reduction in GLT-1 expression, whereas cortical AQP4 immunoreactivity was more intense in the AD group than in the control group. There were two different patterns of GLT-1 and AQP4 expression in the AD group: (i) uneven GLT-1 expression in the neuropil where diffuse but intense AQP4 expression was evident, and (ii) senile plaque-like co-expression of GLT-1 and AQP4. CONCLUSIONS These findings suggest disruption of glutamate/water homoeostasis in the AD brain.
Collapse
Affiliation(s)
- A Hoshi
- Department of Neurology, Fukushima Medical University, Fukushima, Japan.,IMS Shin Katsushika Royal Clinic, Tokyo, Japan
| | - A Tsunoda
- Department of Neurology, Fukushima Medical University, Fukushima, Japan
| | - T Yamamoto
- Department of Neurology, Fukushima Medical University, Fukushima, Japan
| | - M Tada
- Department of Pathology, Brain Research Institute, University of Niigata, Niigata, Japan
| | - A Kakita
- Department of Pathology, Brain Research Institute, University of Niigata, Niigata, Japan
| | - Y Ugawa
- Department of Neurology, Fukushima Medical University, Fukushima, Japan.,Fukushima Global Medical Science Center, Advanced Clinical Research Center, Fukushima Medical University, Fukushima, Japan
| |
Collapse
|
24
|
Rajmohan R, Reddy PH. Amyloid-Beta and Phosphorylated Tau Accumulations Cause Abnormalities at Synapses of Alzheimer's disease Neurons. J Alzheimers Dis 2018; 57:975-999. [PMID: 27567878 DOI: 10.3233/jad-160612] [Citation(s) in RCA: 297] [Impact Index Per Article: 49.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Amyloid-beta (Aβ) and hyperphosphorylated tau are hallmark lesions of Alzheimer's disease (AD). However, the loss of synapses and dysfunctions of neurotransmission are more directly tied to disease severity. The role of these lesions in the pathoetiological progression of the disease remains contested. Biochemical, cellular, molecular, and pathological studies provided several lines of evidence and improved our understanding of how Aβ and hyperphosphorylated tau accumulation may directly harm synapses and alter neurotransmission. In vitro evidence suggests that Aβ and hyperphosphorylated tau have both direct and indirect cytotoxic effects that affect neurotransmission, axonal transport, signaling cascades, organelle function, and immune response in ways that lead to synaptic loss and dysfunctions in neurotransmitter release. Observations in preclinical models and autopsy studies support these findings, suggesting that while the pathoetiology of positive lesions remains elusive, their removal may reduce disease severity and progression. The purpose of this article is to highlight the need for further investigation of the role of tau in disease progression and its interactions with Aβ and neurotransmitters alike.
Collapse
Affiliation(s)
- Ravi Rajmohan
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| | - P Hemachandra Reddy
- Department of Pharmacology and Neuroscience, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Garrison Institute on Aging, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Cell Biology & Biochemistry, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Neurology, Texas Tech University Health Sciences Center, Lubbock, TX, USA.,Department of Speech, Language and Hearing Sciences, Texas Tech University Health Sciences Center, Lubbock, TX, USA
| |
Collapse
|
25
|
Takeda A, Tamano H, Hashimoto W, Kobuchi S, Suzuki H, Murakami T, Tempaku M, Koike Y, Adlard PA, Bush AI. Novel Defense by Metallothionein Induction Against Cognitive Decline: From Amyloid β 1-42-Induced Excess Zn 2+ to Functional Zn 2+ Deficiency. Mol Neurobiol 2018; 55:7775-7788. [PMID: 29460269 DOI: 10.1007/s12035-018-0948-5] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Accepted: 01/31/2018] [Indexed: 02/07/2023]
Abstract
The role of metallothioneins (MTs) in cognitive decline associated with intracellular Zn2+ dysregulation remains unclear. Here, we report that hippocampal MT induction defends cognitive decline, which was induced by amyloid β1-42 (Aβ1-42)-mediated excess Zn2+ and functional Zn2+ deficiency. Excess increase in intracellular Zn2+, which was induced by local injection of Aβ1-42 into the dentate granule cell layer, attenuated in vivo perforant pathway LTP, while the attenuation was rescued by preinjection of MT inducers into the same region. Intraperitoneal injection of dexamethasone, which increased hippocampal MT proteins and blocked Aβ1-42-mediated Zn2+ uptake, but not Aβ1-42 uptake, into dentate granule cells, also rescued Aβ1-42-induced impairment of memory via attenuated LTP. The present study indicates that hippocampal MT induction blocks rapid excess increase in intracellular Zn2+ in dentate granule cells, which originates in Zn2+ released from Aβ1-42, followed by rescuing Aβ1-42-induced cognitive decline. Furthermore, LTP was vulnerable to Aβ1-42 in the aged dentate gyrus, consistent with enhanced Aβ1-42-mediated Zn2+ uptake into aged dentate granule cells, suggesting that Aβ1-42-induced cognitive decline, which is caused by excess intracellular Zn2+, can more frequently occur along with aging. On the other hand, attenuated LTP under functional Zn2+ deficiency in dentate granule cells was also rescued by MT induction. Hippocampal MT induction may rescue cognitive decline under lack of cellular transient changes in functional Zn2+ concentration, while its induction is an attractive defense strategy against Aβ1-42-induced cognitive decline.
Collapse
Affiliation(s)
- Atsushi Takeda
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan.
| | - Haruna Tamano
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Wakana Hashimoto
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Shuhei Kobuchi
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Hiroki Suzuki
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Taku Murakami
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Munekazu Tempaku
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Yuta Koike
- Department of Neurophysiology, School of Pharmaceutical Sciences, University of Shizuoka, 52-1 Yada, Suruga-ku, Shizuoka, 422-8526, Japan
| | - Paul A Adlard
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3052, Australia
| | - Ashley I Bush
- The Florey Institute of Neuroscience and Mental Health, The University of Melbourne, Parkville, VIC, 3052, Australia
| |
Collapse
|
26
|
Caraci F, Nicoletti F, Copani A. Metabotropic glutamate receptors: the potential for therapeutic applications in Alzheimer's disease. Curr Opin Pharmacol 2017; 38:1-7. [PMID: 29278824 DOI: 10.1016/j.coph.2017.12.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2017] [Accepted: 12/11/2017] [Indexed: 02/03/2023]
Abstract
A dysfunction of glutamate signaling is implicated at several levels in the pathogenesis of Alzheimer's disease. Currently, metabotropic glutamate receptors, which have a wide distribution in the central nervous system and activate a multitude of cell signaling pathways, are pursued as targets for therapeutic intervention in Alzheimer's disease. Research is still limited, but results underscore the relevance of ongoing studies. Here we discuss the latest updates regarding metabotropic glutamate receptors and their role in Alzheimer's disease, as well as promising metabotropic glutamate receptor ligands that have been investigated in preclinical models of Alzheimer's disease.
Collapse
Affiliation(s)
- Filippo Caraci
- Department of Drug Sciences, University of Catania, Catania, Italy; Oasi Institute for Research on Mental Retardation and Brain Aging (IRCCS), Troina, Italy
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, University of Rome Sapienza, Rome, Italy; Neuromed (IRCCS), Pozzilli, Italy
| | - Agata Copani
- Department of Drug Sciences, University of Catania, Catania, Italy; Institute of Biostructure and Bioimaging, National Research Council, Catania, Italy.
| |
Collapse
|
27
|
Computer-Aided Drug Design Approaches to Study Key Therapeutic Targets in Alzheimer’s Disease. ACTA ACUST UNITED AC 2017. [DOI: 10.1007/978-1-4939-7404-7_3] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
|
28
|
Amyloid-beta neurotoxicity and clearance are both regulated by glial group II metabotropic glutamate receptors. Neuropharmacology 2017; 123:274-286. [DOI: 10.1016/j.neuropharm.2017.05.008] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2017] [Revised: 05/05/2017] [Accepted: 05/07/2017] [Indexed: 11/20/2022]
|
29
|
Extracellular Zn 2+ Is Essential for Amyloid β 1-42-Induced Cognitive Decline in the Normal Brain and Its Rescue. J Neurosci 2017; 37:7253-7262. [PMID: 28652412 DOI: 10.1523/jneurosci.0954-17.2017] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2017] [Revised: 06/12/2017] [Accepted: 06/19/2017] [Indexed: 12/23/2022] Open
Abstract
Brain Aβ1-42 accumulation is considered an upstream event in pathogenesis of Alzheimer's disease. However, accumulating evidence indicates that other neurochemical changes potentiate the toxicity of this constitutively generated peptide. Here we report that the interaction of Aβ1-42 with extracellular Zn2+ is essential for in vivo rapid uptake of Aβ1-42 and Zn2+ into dentate granule cells in the normal rat hippocampus. The uptake of both Aβ1-42 and Zn2+ was blocked by CaEDTA, an extracellular Zn2+ chelator, and by Cd2+, a metal that displaces Zn2+ for Aβ1-42 binding. In vivo perforant pathway LTP was unaffected by perfusion with 1000 nm Aβ1-42 in ACSF without Zn2+ However, LTP was attenuated under preperfusion with 5 nm Aβ1-42 in ACSF containing 10 nm Zn2+, recapitulating the concentration of extracellular Zn2+, but not with 5 nm Aβ1-40 in ACSF containing 10 nm Zn2+ Aβ1-40 and Zn2+ were not taken up into dentate granule cells under these conditions, consistent with lower affinity of Aβ1-40 for Zn2+ than Aβ1-42 Aβ1-42-induced attenuation of LTP was rescued by both CaEDTA and CdCl2, and was observed even with 500 pm Aβ1-42 Aβ1-42 injected into the dentate granule cell layer of rats induced a rapid memory disturbance that was also rescued by coinjection of CdCl2 The present study supports blocking the formation of Zn-Aβ1-42 in the extracellular compartment as an effective preventive strategy for Alzheimer's disease.SIGNIFICANCE STATEMENT Short-term memory loss occurs in normal elderly and increases in the predementia stage of Alzheimer's disease (AD). Amyloid-β1-42 (Aβ1-42), a possible causing peptide in AD, is bound to Zn2+ in the extracellular compartment in the hippocampus induced short-term memory loss in the normal rat brain, suggesting that extracellular Zn2+ is essential for Aβ1-42-induced short-term memory loss. The evidence is important to find an effective preventive strategy for AD, which is blocking the formation of Zn-Aβ1-42 in the extracellular compartment.
Collapse
|
30
|
Maesako M, Horlacher J, Zoltowska KM, Kastanenka KV, Kara E, Svirsky S, Keller LJ, Li X, Hyman BT, Bacskai BJ, Berezovska O. Pathogenic PS1 phosphorylation at Ser367. eLife 2017; 6. [PMID: 28132667 PMCID: PMC5279945 DOI: 10.7554/elife.19720] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2016] [Accepted: 01/05/2017] [Indexed: 11/13/2022] Open
Abstract
The high levels of serine (S) and threonine (T) residues within the Presenilin 1 (PS1) N-terminus and in the large hydrophilic loop region suggest that the enzymatic function of PS1/γ-secretase can be modulated by its ‘phosphorylated’ and ‘dephosphorylated’ states. However, the functional outcome of PS1 phosphorylation and its significance for Alzheimer’s disease (AD) pathogenesis is poorly understood. Here, comprehensive analysis using FRET-based imaging reveals that activity-driven and Protein Kinase A-mediated PS1 phosphorylation at three domains (domain 1: T74, domain 2: S310 and S313, domain 3: S365, S366, and S367), with S367 being critical, is responsible for the PS1 pathogenic ‘closed’ conformation, and resulting increase in the Aβ42/40 ratio. Moreover, we have established novel imaging assays for monitoring PS1 conformation in vivo, and report that PS1 phosphorylation induces the pathogenic conformational shift in the living mouse brain. These phosphorylation sites represent potential new targets for AD treatment. DOI:http://dx.doi.org/10.7554/eLife.19720.001 Alzheimer’s disease is a widely recognised disorder caused by the progressive deterioration and death of brain cells. A key feature of the disease is the formation of structures called plaques in the brain. Plaques occur when many copies of a molecule known as amyloid beta stick together outside of the brain cells. Healthy brains also produce amyloid beta but it is in a different form, which cannot form plaques. One in twenty people with Alzheimer’s disease have a family history of the disease. Of these, many are linked to changes in a gene that produces a protein called Presenilin 1 (or PS1 for short). Cells need PS1 to make amyloid beta and the altered versions of PS1 produce the type of amyloid beta that causes Alzheimer’s disease. Yet, in cases that do not run in families, the gene for PS1 is unchanged but the PS1 protein still produces the form of amyloid beta that is linked to Alzheimer’s disease. Maesako, Horlacher et al. wanted to find out how seemingly healthy PS1 proteins can be made to produce plaque-forming amyloid betas. Studies of PS1 from mice revealed that small chemical modifications, called phosphate groups, could be attached to PS1 in a process called phosphorylation. Modified PS1 proteins produce harmful amyloid betas and removing the modifications was enough to make PS1 behave normally again. Maesako, Horlacher et al. found three points in the PS1 protein where phosphorylation could change the behaviour of the protein, the most important one is a site called Ser367. Further investigation showed that an enzyme called Protein Kinase A (PKA) phosphorylates PS1; this enzyme is also able to attach phosphate groups to many different proteins. Maesako, Horlacher et al. went on to show that PS1 is phosphorylated in samples from people with Alzheimer’s disease, suggesting that this is a plausible cause for some cases of the disease. Finding a way to prevent phosphorylation or remove phosphate groups from PS1 could be the first step towards treating these cases of Alzheimer’s disease. DOI:http://dx.doi.org/10.7554/eLife.19720.002
Collapse
Affiliation(s)
- Masato Maesako
- Alzheimer's Disease Research Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Jana Horlacher
- Alzheimer's Disease Research Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States.,Department of Neurology, University of Ulm, Ulm, Germany
| | - Katarzyna M Zoltowska
- Alzheimer's Disease Research Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Ksenia V Kastanenka
- Alzheimer's Disease Research Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Eleanna Kara
- Alzheimer's Disease Research Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Sarah Svirsky
- Alzheimer's Disease Research Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Laura J Keller
- Alzheimer's Disease Research Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Xuejing Li
- Alzheimer's Disease Research Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Bradley T Hyman
- Alzheimer's Disease Research Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Brian J Bacskai
- Alzheimer's Disease Research Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| | - Oksana Berezovska
- Alzheimer's Disease Research Laboratory, MassGeneral Institute for Neurodegenerative Disease, Massachusetts General Hospital, Harvard Medical School, Charlestown, United States
| |
Collapse
|
31
|
Metabotropic glutamate receptors and neurodegenerative diseases. Pharmacol Res 2017; 115:179-191. [DOI: 10.1016/j.phrs.2016.11.013] [Citation(s) in RCA: 145] [Impact Index Per Article: 20.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Revised: 11/11/2016] [Accepted: 11/15/2016] [Indexed: 12/21/2022]
|
32
|
Lindsley CW, Emmitte KA, Hopkins CR, Bridges TM, Gregory KJ, Niswender CM, Conn PJ. Practical Strategies and Concepts in GPCR Allosteric Modulator Discovery: Recent Advances with Metabotropic Glutamate Receptors. Chem Rev 2016; 116:6707-41. [PMID: 26882314 PMCID: PMC4988345 DOI: 10.1021/acs.chemrev.5b00656] [Citation(s) in RCA: 135] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Allosteric modulation of GPCRs has initiated a new era of basic and translational discovery, filled with therapeutic promise yet fraught with caveats. Allosteric ligands stabilize unique conformations of the GPCR that afford fundamentally new receptors, capable of novel pharmacology, unprecedented subtype selectivity, and unique signal bias. This review provides a comprehensive overview of the basics of GPCR allosteric pharmacology, medicinal chemistry, drug metabolism, and validated approaches to address each of the major challenges and caveats. Then, the review narrows focus to highlight recent advances in the discovery of allosteric ligands for metabotropic glutamate receptor subtypes 1-5 and 7 (mGlu1-5,7) highlighting key concepts ("molecular switches", signal bias, heterodimers) and practical solutions to enable the development of tool compounds and clinical candidates. The review closes with a section on late-breaking new advances with allosteric ligands for other GPCRs and emerging data for endogenous allosteric modulators.
Collapse
Affiliation(s)
- Craig W. Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Department of Chemistry, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Kyle A. Emmitte
- Department of Pharmaceutical Sciences, UNT System College of Pharmacy, University of North Texas Health Science Center, 3500 Camp Bowie Boulevard, Fort Worth, Texas 76107, United States
| | - Corey R. Hopkins
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Thomas M. Bridges
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - Karen J. Gregory
- Drug Discovery Biology, Monash Institute of Pharmaceutical Sciences, Monash University, 381 Royal Parade, Parkville VIC 3052, Australia
| | - Colleen M. Niswender
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| | - P. Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
- Vanderbilt Kennedy Center, Vanderbilt University School of Medicine, Nashville, Tennessee 37232, United States
| |
Collapse
|
33
|
Yan R, Fan Q, Zhou J, Vassar R. Inhibiting BACE1 to reverse synaptic dysfunctions in Alzheimer's disease. Neurosci Biobehav Rev 2016; 65:326-40. [PMID: 27044452 PMCID: PMC4856578 DOI: 10.1016/j.neubiorev.2016.03.025] [Citation(s) in RCA: 53] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2015] [Revised: 03/25/2016] [Accepted: 03/29/2016] [Indexed: 12/21/2022]
Abstract
Over the past two decades, many studies have identified significant contributions of toxic β-amyloid peptides (Aβ) to the etiology of Alzheimer's disease (AD), which is the most common age-dependent neurodegenerative disease. AD is also recognized as a disease of synaptic failure. Aβ, generated by sequential proteolytic cleavages of amyloid precursor protein (APP) by BACE1 and γ-secretase, is one of major culprits that cause this failure. In this review, we summarize current findings on how BACE1-cleaved APP products impact learning and memory through proteins localized on glutamatergic, GABAergic, and dopaminergic synapses. Considering the broad effects of Aβ on all three types of synapses, BACE1 inhibition emerges as a practical approach for ameliorating Aβ-mediated synaptic dysfunctions. Since BACE1 inhibitory drugs are currently in clinical trials, this review also discusses potential complications arising from BACE1 inhibition. We emphasize that the benefits of BACE1 inhibitory drugs will outweigh the concerns.
Collapse
Affiliation(s)
- Riqiang Yan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| | - Qingyuan Fan
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - John Zhou
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA
| | - Robert Vassar
- Department of Neurosciences, Lerner Research Institute, Cleveland Clinic, Cleveland, OH 44195, USA.
| |
Collapse
|
34
|
Bruno V, Caraci F, Copani A, Matrisciano F, Nicoletti F, Battaglia G. The impact of metabotropic glutamate receptors into active neurodegenerative processes: A "dark side" in the development of new symptomatic treatments for neurologic and psychiatric disorders. Neuropharmacology 2016; 115:180-192. [PMID: 27140693 DOI: 10.1016/j.neuropharm.2016.04.044] [Citation(s) in RCA: 52] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/11/2016] [Revised: 04/22/2016] [Accepted: 04/28/2016] [Indexed: 12/17/2022]
Abstract
Metabotropic glutamate (mGlu) receptor ligands are under clinical development for the treatment of CNS disorders with high social and economic burden, such as schizophrenia, major depressive disorder (MDD), and Parkinson's disease (PD), and are promising drug candidates for the treatment of Alzheimer's disease (AD). So far, clinical studies have shown symptomatic effects of mGlu receptor ligands, but it is unknown whether these drugs act as disease modifiers or, at the opposite end, they accelerate disease progression by enhancing neurodegeneration. This is a fundamental issue in the treatment of PD and AD, and is also an emerging theme in the treatment of schizophrenia and MDD, in which neurodegeneration is also present and contribute to disease progression. Moving from in vitro data and preclinical studies, we discuss the potential impact of drugs targeting mGlu2, mGlu3, mGlu4 and mGlu5 receptor ligands on active neurodegeneration associated with AD, PD, schizophrenia, and MDD. We wish to highlight that our final comments on the best drug candidates are not influenced by commercial interests or by previous or ongoing collaborations with drug companies. This article is part of the Special Issue entitled 'Metabotropic Glutamate Receptors, 5 years on'.
Collapse
Affiliation(s)
- Valeria Bruno
- Department of Physiology and Pharmacology, University Sapienza, 00185 Rome, Italy; I.R.C.C.S. Neuromed, 86077 Pozzilli, Italy.
| | - Filippo Caraci
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; I.R.C.C.S. Associazione Oasi Maria S.S., Institute for Research on Mental Retardation and Brain Aging, 94018 Troina, Italy
| | - Agata Copani
- Department of Drug Sciences, University of Catania, 95125 Catania, Italy; National Research Council, Institute of Biostructure and Bioimaging (IBB-CNR), 95126 Catania, Italy
| | - Francesco Matrisciano
- Department of Psychiatry and Behavioral Sciences, Northwestern Feinberg School of Medicine, Chicago, USA
| | - Ferdinando Nicoletti
- Department of Physiology and Pharmacology, University Sapienza, 00185 Rome, Italy; I.R.C.C.S. Neuromed, 86077 Pozzilli, Italy
| | | |
Collapse
|
35
|
Kuzuya A, Zoltowska KM, Post KL, Arimon M, Li X, Svirsky S, Maesako M, Muzikansky A, Gautam V, Kovacs D, Hyman BT, Berezovska O. Identification of the novel activity-driven interaction between synaptotagmin 1 and presenilin 1 links calcium, synapse, and amyloid beta. BMC Biol 2016; 14:25. [PMID: 27036734 PMCID: PMC4818459 DOI: 10.1186/s12915-016-0248-3] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2016] [Accepted: 03/18/2016] [Indexed: 12/20/2022] Open
Abstract
Background Synaptic loss strongly correlates with memory deterioration. Local accumulation of amyloid β (Aβ) peptide, and neurotoxic Aβ42 in particular, due to abnormal neuronal activity may underlie synaptic dysfunction, neurodegeneration, and memory impairments. To gain an insight into molecular events underlying neuronal activity-regulated Aβ production at the synapse, we explored functional outcomes of the newly discovered calcium-dependent interaction between Alzheimer’s disease-associated presenilin 1 (PS1)/γ-secretase and synaptic vesicle proteins. Results Mass spectrometry screen of mouse brain lysates identified synaptotagmin 1 (Syt1) as a novel synapse-specific PS1-binding partner that shows Ca2+-dependent PS1 binding profiles in vitro and in vivo. We found that Aβ level, and more critically, conformation of the PS1 and the Aβ42/40 ratio, are affected by Syt1 overexpression or knockdown, indicating that Syt1 and its interaction with PS1 might regulate Aβ production at the synapse. Moreover, β-secretase 1 (BACE1) stability, β- and γ-secretase activity, as well as intracellular compartmentalization of PS1 and BACE1, but not of amyloid precursor protein (APP), nicastrin (Nct), presenilin enhancer 2 (Pen-2), or synaptophysin (Syp) were altered in the absence of Syt1, suggesting a selective effect of Syt1 on PS1 and BACE1 trafficking. Conclusions Our findings identify Syt1 as a novel Ca2+-sensitive PS1 modulator that could regulate synaptic Aβ, opening avenues for novel and selective synapse targeting therapeutic strategies. Electronic supplementary material The online version of this article (doi:10.1186/s12915-016-0248-3) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Akira Kuzuya
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Katarzyna M Zoltowska
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Kathryn L Post
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Muriel Arimon
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Xuejing Li
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Sarah Svirsky
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Masato Maesako
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Alona Muzikansky
- MGH Biostatistics Center, Massachusetts General Hospital, Boston, MA, 02114, USA
| | - Vivek Gautam
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Dora Kovacs
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Bradley T Hyman
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA
| | - Oksana Berezovska
- MassGeneral Institute for Neurodegenerative Disease, Department of Neurology, Massachusetts General Hospital, Harvard Medical School, Charlestown, MA, 02129, USA.
| |
Collapse
|
36
|
Zhao J, Deng Y, Jiang Z, Qing H. G Protein-Coupled Receptors (GPCRs) in Alzheimer's Disease: A Focus on BACE1 Related GPCRs. Front Aging Neurosci 2016; 8:58. [PMID: 27047374 PMCID: PMC4805599 DOI: 10.3389/fnagi.2016.00058] [Citation(s) in RCA: 60] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/07/2016] [Accepted: 03/08/2016] [Indexed: 12/22/2022] Open
Abstract
The G protein coupled receptors (GPCRs) have been considered as one of the largest families of validated drug targets, which involve in almost overall physiological functions and pathological processes. Meanwhile, Alzheimer’s disease (AD), the most common type of dementia, affects thinking, learning, memory and behavior of elderly people, that has become the hotspot nowadays for its increasing risks and incurability. The above fields have been intensively studied, and the link between the two has been demonstrated, whereas the way how GPCRs perturb AD progress are yet to be further explored given their complexities. In this review, we summarized recent progress regarding the GPCRs interacted with β-site APP cleaving enzyme 1 (BACE1), a key secretase in AD pathogenesis. Then we discussed the current findings on the regulatory roles of GPCRs on BACE1, and the possibility for pharmaceutical treatment of AD patients by the allosteric modulators and biased ligands of GPCRs. We hope this review can provide new insights into the understanding of mechanistic link between GPCRs and BACE1, and highlight the potential of GPCRs as therapeutic target for AD.
Collapse
Affiliation(s)
- Juan Zhao
- School of Life Science, Beijing Institute of Technology Beijing, China
| | - Yulin Deng
- School of Life Science, Beijing Institute of Technology Beijing, China
| | - Zhaotan Jiang
- School of Physics, Beijing Institute of Technology Beijing, China
| | - Hong Qing
- School of Life Science, Beijing Institute of Technology Beijing, China
| |
Collapse
|
37
|
Felts AS, Rodriguez AL, Smith KA, Engers JL, Morrison RD, Byers FW, Blobaum AL, Locuson CW, Chang S, Venable DF, Niswender CM, Daniels JS, Conn PJ, Lindsley CW, Emmitte KA. Design of 4-Oxo-1-aryl-1,4-dihydroquinoline-3-carboxamides as Selective Negative Allosteric Modulators of Metabotropic Glutamate Receptor Subtype 2. J Med Chem 2015; 58:9027-40. [PMID: 26524606 DOI: 10.1021/acs.jmedchem.5b01371] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
Both orthosteric and allosteric antagonists of the group II metabotropic glutamate receptors (mGlus) have been used to establish a link between mGlu2/3 inhibition and a variety of CNS diseases and disorders. Though these tools typically have good selectivity for mGlu2/3 versus the remaining six members of the mGlu family, compounds that are selective for only one of the individual group II mGlus have proved elusive. Herein we report on the discovery of a potent and highly selective mGlu2 negative allosteric modulator 58 (VU6001192) from a series of 4-oxo-1-aryl-1,4-dihydroquinoline-3-carboxamides. The concept for the design of this series centered on morphing a quinoline series recently disclosed in the patent literature into a chemotype previously used for the preparation of muscarinic acetylcholine receptor subtype 1 positive allosteric modulators. Compound 58 exhibits a favorable profile and will be a useful tool for understanding the biological implications of selective inhibition of mGlu2 in the CNS.
Collapse
Affiliation(s)
- Andrew S Felts
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Alice L Rodriguez
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Katrina A Smith
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Julie L Engers
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Ryan D Morrison
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Frank W Byers
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Anna L Blobaum
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Charles W Locuson
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Sichen Chang
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Daryl F Venable
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Colleen M Niswender
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - J Scott Daniels
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - P Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Craig W Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States.,Department of Chemistry, Vanderbilt University , Nashville, Tennessee 37232, United States
| | - Kyle A Emmitte
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States.,Department of Chemistry, Vanderbilt University , Nashville, Tennessee 37232, United States
| |
Collapse
|
38
|
Engers JL, Rodriguez AL, Konkol LC, Morrison RD, Thompson AD, Byers FW, Blobaum AL, Chang S, Venable DF, Loch MT, Niswender CM, Daniels JS, Jones CK, Conn PJ, Lindsley CW, Emmitte KA. Discovery of a Selective and CNS Penetrant Negative Allosteric Modulator of Metabotropic Glutamate Receptor Subtype 3 with Antidepressant and Anxiolytic Activity in Rodents. J Med Chem 2015; 58:7485-500. [PMID: 26335039 DOI: 10.1021/acs.jmedchem.5b01005] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Previous preclinical work has demonstrated the therapeutic potential of antagonists of the group II metabotropic glutamate receptors (mGlus). Still, compounds that are selective for the individual group II mGlus (mGlu2 and mGlu3) have been scarce. There remains a need for such compounds with the balance of properties suitable for convenient use in a wide array of rodent behavioral studies. We describe here the discovery of a selective mGlu3 NAM 106 (VU0650786) suitable for in vivo work. Compound 106 is a member of a series of 5-aryl-6,7-dihydropyrazolo[1,5-a]pyrazine-4(5H)-one compounds originally identified as a mGlu5 positive allosteric modulator (PAM) chemotype. Its suitability for use in rodent behavioral models has been established by extensive in vivo PK studies, and the behavioral experiments presented here with compound 106 represent the first examples in which an mGlu3 NAM has demonstrated efficacy in models where prior efficacy had previously been noted with nonselective group II antagonists.
Collapse
Affiliation(s)
- Julie L Engers
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Alice L Rodriguez
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Leah C Konkol
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Ryan D Morrison
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Analisa D Thompson
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Frank W Byers
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Anna L Blobaum
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Sichen Chang
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Daryl F Venable
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Matthew T Loch
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Colleen M Niswender
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - J Scott Daniels
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Carrie K Jones
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - P Jeffrey Conn
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States
| | - Craig W Lindsley
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States.,Department of Chemistry, Vanderbilt University , Nashville, Tennessee 37232, United States
| | - Kyle A Emmitte
- Vanderbilt Center for Neuroscience Drug Discovery, Department of Pharmacology, Vanderbilt University Medical Center , Nashville, Tennessee 37232, United States.,Department of Chemistry, Vanderbilt University , Nashville, Tennessee 37232, United States
| |
Collapse
|
39
|
Takahashi K, Foster JB, Lin CLG. Glutamate transporter EAAT2: regulation, function, and potential as a therapeutic target for neurological and psychiatric disease. Cell Mol Life Sci 2015; 72:3489-506. [PMID: 26033496 PMCID: PMC11113985 DOI: 10.1007/s00018-015-1937-8] [Citation(s) in RCA: 121] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2015] [Revised: 05/22/2015] [Accepted: 05/26/2015] [Indexed: 12/12/2022]
Abstract
Glutamate is the predominant excitatory neurotransmitter in the central nervous system. Excitatory amino acid transporter 2 (EAAT2) is primarily responsible for clearance of extracellular glutamate to prevent neuronal excitotoxicity and hyperexcitability. EAAT2 plays a critical role in regulation of synaptic activity and plasticity. In addition, EAAT2 has been implicated in the pathogenesis of many central nervous system disorders. In this review, we summarize current understanding of EAAT2, including structure, pharmacology, physiology, and functions, as well as disease relevancy, such as in stroke, Parkinson's disease, epilepsy, amyotrophic lateral sclerosis, Alzheimer's disease, major depressive disorder, and addiction. A large number of studies have demonstrated that up-regulation of EAAT2 protein provides significant beneficial effects in many disease models suggesting EAAT2 activation is a promising therapeutic approach. Several EAAT2 activators have been identified. Further understanding of EAAT2 regulatory mechanisms could improve development of drug-like compounds that spatiotemporally regulate EAAT2.
Collapse
Affiliation(s)
- Kou Takahashi
- Department of Neuroscience, The Ohio State University, 333 West 10th Avenue, Columbus, OH 43210 USA
| | - Joshua B. Foster
- Department of Neuroscience, The Ohio State University, 333 West 10th Avenue, Columbus, OH 43210 USA
| | - Chien-Liang Glenn Lin
- Department of Neuroscience, The Ohio State University, 333 West 10th Avenue, Columbus, OH 43210 USA
| |
Collapse
|
40
|
Takahashi K, Kong Q, Lin Y, Stouffer N, Schulte DA, Lai L, Liu Q, Chang LC, Dominguez S, Xing X, Cuny GD, Hodgetts KJ, Glicksman MA, Lin CLG. Restored glial glutamate transporter EAAT2 function as a potential therapeutic approach for Alzheimer's disease. ACTA ACUST UNITED AC 2015; 212:319-32. [PMID: 25711212 PMCID: PMC4354363 DOI: 10.1084/jem.20140413] [Citation(s) in RCA: 103] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023]
Abstract
Takahashi et al. demonstrate that restoring glial glutamate transporter EAAT2 function improves cognitive functions and synaptic integrity while reducing amyloid plaques in a sustained fashion after treatment cessation. Glutamatergic systems play a critical role in cognitive functions and are known to be defective in Alzheimer’s disease (AD) patients. Previous literature has indicated that glial glutamate transporter EAAT2 plays an essential role in cognitive functions and that loss of EAAT2 protein is a common phenomenon observed in AD patients and animal models. In the current study, we investigated whether restored EAAT2 protein and function could benefit cognitive functions and pathology in APPSw,Ind mice, an animal model of AD. A transgenic mouse approach via crossing EAAT2 transgenic mice with APPSw,Ind. mice and a pharmacological approach using a novel EAAT2 translational activator, LDN/OSU-0212320, were conducted. Findings from both approaches demonstrated that restored EAAT2 protein function significantly improved cognitive functions, restored synaptic integrity, and reduced amyloid plaques. Importantly, the observed benefits were sustained one month after compound treatment cessation, suggesting that EAAT2 is a potential disease modifier with therapeutic potential for AD.
Collapse
Affiliation(s)
- Kou Takahashi
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210
| | - Qiongman Kong
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210
| | - Yuchen Lin
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210
| | - Nathan Stouffer
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210
| | - Delanie A Schulte
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210
| | - Liching Lai
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210
| | - Qibing Liu
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210
| | - Ling-Chu Chang
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210
| | - Sky Dominguez
- Department of Neuroscience, The Ohio State University, Columbus, OH 43210
| | - Xuechao Xing
- Laboratory for Drug Discovery in Neurodegeneration, Harvard NeuroDiscovery Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Gregory D Cuny
- Laboratory for Drug Discovery in Neurodegeneration, Harvard NeuroDiscovery Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115 Department of Pharmacological and Pharmaceutical Sciences, University of Houston College of Pharmacy, Houston, TX 77004
| | - Kevin J Hodgetts
- Laboratory for Drug Discovery in Neurodegeneration, Harvard NeuroDiscovery Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | - Marcie A Glicksman
- Laboratory for Drug Discovery in Neurodegeneration, Harvard NeuroDiscovery Center, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115
| | | |
Collapse
|
41
|
Role of Group II Metabotropic Glutamate Receptors (mGluR2/3) Blockade on Long-Term Potentiation in the Dentate Gyrus Region of Hippocampus in Rats Fed with High-Fat Diet. Neurochem Res 2015; 40:811-7. [DOI: 10.1007/s11064-015-1531-3] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2014] [Revised: 01/24/2015] [Accepted: 01/29/2015] [Indexed: 10/24/2022]
|
42
|
Chumakov I, Nabirotchkin S, Cholet N, Milet A, Boucard A, Toulorge D, Pereira Y, Graudens E, Traoré S, Foucquier J, Guedj M, Vial E, Callizot N, Steinschneider R, Maurice T, Bertrand V, Scart-Grès C, Hajj R, Cohen D. Combining two repurposed drugs as a promising approach for Alzheimer's disease therapy. Sci Rep 2015; 5:7608. [PMID: 25566747 PMCID: PMC5378993 DOI: 10.1038/srep07608] [Citation(s) in RCA: 46] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2014] [Accepted: 11/19/2014] [Indexed: 02/08/2023] Open
Abstract
Alzheimer disease (AD) represents a major medical problem where mono-therapeutic interventions demonstrated only a limited efficacy so far. We explored the possibility of developing a combinational therapy that might prevent the degradation of neuronal and endothelial structures in this disease. We argued that the distorted balance between excitatory (glutamate) and inhibitory (GABA/glycine) systems constitutes a therapeutic target for such intervention. We found that a combination of two approved drugs – acamprosate and baclofen – synergistically protected neurons and endothelial structures in vitro against amyloid-beta (Aβ) oligomers. The neuroprotective effects of these drugs were mediated by modulation of targets in GABA/glycinergic and glutamatergic pathways. In vivo, the combination alleviated cognitive deficits in the acute Aβ25–35 peptide injection model and in the mouse mutant APP transgenic model. Several patterns altered in AD were also synergistically normalised. Our results open up the possibility for a promising therapeutic approach for AD by combining repurposed drugs.
Collapse
Affiliation(s)
- Ilya Chumakov
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | | | - Nathalie Cholet
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Aude Milet
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Aurélie Boucard
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Damien Toulorge
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Yannick Pereira
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Esther Graudens
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Sory Traoré
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Julie Foucquier
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Mickael Guedj
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Emmanuel Vial
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | | | | | - Tangui Maurice
- 1] Université de Montpellier 2, 34095 Montpellier, France; Inserm, U710, 34095 Montpellier, France; EPHE, 75017 Paris, France [2] Amylgen, 2196 bd de la Lironde, 34980 Montferrier-sur-Lez, France
| | - Viviane Bertrand
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | | | - Rodolphe Hajj
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| | - Daniel Cohen
- Pharnext, 11 rue des Peupliers, 92130 Issy-Les-Moulineaux, France
| |
Collapse
|
43
|
Proneurogenic Group II mGluR antagonist improves learning and reduces anxiety in Alzheimer Aβ oligomer mouse. Mol Psychiatry 2014; 19:1235-42. [PMID: 25113378 PMCID: PMC4217144 DOI: 10.1038/mp.2014.87] [Citation(s) in RCA: 47] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/19/2014] [Revised: 06/02/2014] [Accepted: 06/06/2014] [Indexed: 01/30/2023]
Abstract
Proneurogenic compounds have recently shown promise in some mouse models of Alzheimer's pathology. Antagonists at Group II metabotropic glutamate receptors (Group II mGluR: mGlu2, mGlu3) are reported to stimulate neurogenesis. Agonists at those receptors trigger γ-secretase-inhibitor-sensitive biogenesis of Aβ42 peptides from isolated synaptic terminals, which is selectively suppressed by antagonist pretreatment. We have assessed the therapeutic potential of chronic pharmacological inhibition of Group II mGluR in Dutch APP (Alzheimer's amyloid precursor protein E693Q) transgenic mice that accumulate Dutch amyloid-β (Aβ) oligomers but never develop Aβ plaques. BCI-838 is a clinically well-tolerated, orally bioavailable, investigational prodrug that delivers to the brain BCI-632, the active Group II mGluR antagonist metabolite. Dutch Aβ-oligomer-forming APP transgenic mice (APP E693Q) were dosed with BCI-838 for 3 months. Chronic treatment with BCI-838 was associated with reversal of transgene-related amnestic behavior, reduction in anxiety, reduction in levels of brain Aβ monomers and oligomers, and stimulation of hippocampal neurogenesis. Group II mGluR inhibition may offer a unique package of relevant properties as an Alzheimer's disease therapeutic or prophylactic by providing both attenuation of neuropathology and stimulation of repair.
Collapse
|
44
|
Cynis H, Funkelstein L, Toneff T, Mosier C, Ziegler M, Koch B, Demuth HU, Hook V. Pyroglutamate-amyloid-β and glutaminyl cyclase are colocalized with amyloid-β in secretory vesicles and undergo activity-dependent, regulated secretion. NEURODEGENER DIS 2014; 14:85-97. [PMID: 24943989 DOI: 10.1159/000358430] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2013] [Accepted: 01/07/2014] [Indexed: 12/14/2022] Open
Abstract
BACKGROUND AND AIMS N-truncated pyroglutamate (pGlu)-amyloid-β [Aβ(3-40/42)] peptides are key components that promote Aβ peptide accumulation, leading to neurodegeneration and memory loss in Alzheimer's disease. Because Aβ deposition in the brain occurs in an activity-dependent manner, it is important to define the subcellular organelle for pGlu-Aβ(3-40/42) production by glutaminyl cyclase (QC) and their colocalization with full-length Aβ(1-40/42) peptides for activity-dependent, regulated secretion. Therefore, the objective of this study was to investigate the hypothesis that pGlu-Aβ and QC are colocalized with Aβ in dense-core secretory vesicles (DCSV) for activity-dependent secretion with neurotransmitters. METHODS Purified DCSV were assessed for pGlu-Aβ(3-40/42), Aβ(1-40/42), QC, and neurotransmitter secretion. Neuron-like chromaffin cells were analyzed for cosecretion of pGlu-Aβ, QC, Aβ, and neuropeptides. The cells were treated with a QC inhibitor, and pGlu-Aβ production was measured. Human neuroblastoma cells were also examined for pGlu-Aβ and QC secretion. RESULTS Isolated DCSV contain pGlu-Aβ(3-40/42), QC, and Aβ(1-40/42) with neuropeptide and catecholamine neurotransmitters. Cellular pGlu-Aβ and QC undergo activity-dependent cosecretion with Aβ and enkephalin and galanin neurotransmitters. The QC inhibitor decreased the level of secreted pGlu-Aβ. The human neuroblastoma cells displayed regulated secretion of pGlu-Aβ that was colocalized with QC. CONCLUSIONS pGlu-Aβ and QC are present with Aβ in DCSV and undergo activity-dependent, regulated cosecretion with neurotransmitters.
Collapse
|
45
|
Yin S, Niswender CM. Progress toward advanced understanding of metabotropic glutamate receptors: structure, signaling and therapeutic indications. Cell Signal 2014; 26:2284-97. [PMID: 24793301 DOI: 10.1016/j.cellsig.2014.04.022] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2014] [Accepted: 04/27/2014] [Indexed: 12/24/2022]
Abstract
The metabotropic glutamate (mGlu) receptors are a group of Class C seven-transmembrane spanning/G protein-coupled receptors (7TMRs/GPCRs). These receptors are activated by glutamate, one of the standard amino acids and the major excitatory neurotransmitter. By activating G protein-dependent and non-G protein-dependent signaling pathways, mGlus modulate glutamatergic transmission both in the periphery and throughout the central nervous system. Since the discovery of the first mGlu receptor, and especially during the last decade, a great deal of progress has been made in understanding the signaling, structure, pharmacological manipulation and therapeutic indications of the 8 mGlu members.
Collapse
Affiliation(s)
- Shen Yin
- Department of Pharmacology, Vanderbilt University Medical School, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical School, Nashville, TN 37232, USA
| | - Colleen M Niswender
- Department of Pharmacology, Vanderbilt University Medical School, Nashville, TN 37232, USA; Vanderbilt Center for Neuroscience Drug Discovery, Vanderbilt University Medical School, Nashville, TN 37232, USA.
| |
Collapse
|
46
|
Gandy S. Alzheimer's disease: new data highlight nonneuronal cell types and the necessity for presymptomatic prevention strategies. Biol Psychiatry 2014; 75:553-7. [PMID: 24373429 PMCID: PMC4013678 DOI: 10.1016/j.biopsych.2013.11.019] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/28/2013] [Revised: 10/23/2013] [Accepted: 11/22/2013] [Indexed: 02/04/2023]
Abstract
Despite compelling genetic evidence indicating that cerebral amyloidosis can be, at least sometimes, the primary cause of Alzheimer's disease (AD), clinical trials for symptomatic AD with amyloid-reducing agents have succeeded at target engagement but failed to cause clinical benefit. In a landmark shift, the U.S. Food and Drug Administration now proposes to approve prophylaxis that alters the trajectory of what is now believed to be typical AD biomarker evolution. The first prevention trials are now beginning in patients with genetic guarantees for or high genetic risks for AD. The expectation is that clues to their outcomes will begin to emerge from these trials in approximately 2018. In the meantime, new strategies point to nonneuronal cells and to system pathology. A review of the current state of the art of AD science follows herein.
Collapse
Affiliation(s)
- Sam Gandy
- Departments of Neurology and Psychiatry, and the Mount Sinai Alzheimer's Disease Research Center, Icahn School of Medicine at Mount Sinai, and James J. Peters VA Medical Center, New York, New York.
| |
Collapse
|
47
|
Durand D, Carniglia L, Beauquis J, Caruso C, Saravia F, Lasaga M. Astroglial mGlu3 receptors promote alpha-secretase-mediated amyloid precursor protein cleavage. Neuropharmacology 2014; 79:180-9. [DOI: 10.1016/j.neuropharm.2013.11.015] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2013] [Revised: 09/26/2013] [Accepted: 11/20/2013] [Indexed: 12/21/2022]
|
48
|
Lundgren JL, Ahmed S, Winblad B, Gouras GK, Tjernberg LO, Frykman S. Activity-independent release of the amyloid β-peptide from rat brain nerve terminals. Neurosci Lett 2014; 566:125-30. [PMID: 24602978 DOI: 10.1016/j.neulet.2014.02.050] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2013] [Revised: 02/10/2014] [Accepted: 02/23/2014] [Indexed: 01/21/2023]
Abstract
Synaptic degeneration is one of the earliest hallmarks of Alzheimer disease. The molecular mechanism underlying this degeneration is not fully elucidated but one key player appears to be the synaptotoxic amyloid β-peptide (Aβ). The exact localization of the production of Aβ and the mechanisms whereby Aβ is released remain elusive. We have earlier shown that Aβ can be produced in crude synaptic vesicle fractions and it has been reported that increased synaptic activity results in increased secreted but decreased intracellular Aβ levels. Therefore, we considered whether Aβ could be produced in synaptic vesicles and/or released through the same mechanisms as neurotransmitters in synaptic vesicle exocytosis. Small amounts of Aβ were found to be produced in pure synaptic vesicle preparations. We also studied the release of glutamate and Aβ from rat cortical nerve terminals (synaptosomes). We found that large amounts of Aβ were secreted from non-stimulated synaptosomes, from which glutamate was not released. On the contrary, we could not detect any differences in Aβ release between non-stimulated synaptosomes and synaptosomes stimulated with KCl or 4-aminopyridine, whereas glutamate release was readily inducible in this system. To conclude, our results indicate that the major release mechanism of Aβ from isolated nerve terminals differs from the synaptic release of glutamate and that the activity-dependent increase of secreted Aβ, reported by several groups using intact cells, is likely dependent on post-synaptic events, trafficking and/or protein synthesis mechanisms.
Collapse
Affiliation(s)
- Jolanta L Lundgren
- Karolinska Institutet, Department of Neurobiology, Care Science and Society, Center for Alzheimer Research, Novum level 5, 141 86 Stockholm, Sweden
| | - Saheeb Ahmed
- European Neuroscience Institute, Grisebachstrasse 5, 37077 Göttingen, Germany
| | - Bengt Winblad
- Karolinska Institutet, Department of Neurobiology, Care Science and Society, Center for Alzheimer Research, Novum level 5, 141 86 Stockholm, Sweden
| | - Gunnar K Gouras
- Lund University, Department of Experimental Medical Science, Experimental Dementia Research Unit, Sölveg 19, BMC B12, 221 84 Lund, Sweden
| | - Lars O Tjernberg
- Karolinska Institutet, Department of Neurobiology, Care Science and Society, Center for Alzheimer Research, Novum level 5, 141 86 Stockholm, Sweden
| | - Susanne Frykman
- Karolinska Institutet, Department of Neurobiology, Care Science and Society, Center for Alzheimer Research, Novum level 5, 141 86 Stockholm, Sweden.
| |
Collapse
|
49
|
Holland JP, Liang SH, Rotstein BH, Collier TL, Stephenson NA, Greguric I, Vasdev N. Alternative approaches for PET radiotracer development in Alzheimer's disease: imaging beyond plaque. J Labelled Comp Radiopharm 2013; 57:323-31. [PMID: 24327420 DOI: 10.1002/jlcr.3158] [Citation(s) in RCA: 36] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2013] [Accepted: 10/29/2013] [Indexed: 12/18/2022]
Abstract
Alzheimer's disease (AD) and related dementias show increasing clinical prevalence, yet our understanding of the etiology and pathobiology of disease-related neurodegeneration remains limited. In this regard, noninvasive imaging with radiotracers for positron emission tomography (PET) presents a unique tool for quantifying spatial and temporal changes in characteristic biological markers of brain disease and for assessing potential drug efficacy. PET radiotracers targeting different protein markers are being developed to address questions pertaining to the molecular and/or genetic heterogeneity of AD and related dementias. For example, radiotracers including [(11) C]-PiB and [(18) F]-AV-45 (Florbetapir) are being used to measure the density of Aβ-plaques in AD patients and to interrogate the biological mechanisms of disease initiation and progression. Our focus is on the development of novel PET imaging agents, targeting proteins beyond Aβ-plaques, which can be used to investigate the broader mechanism of AD pathogenesis. Here, we present the chemical basis of various radiotracers which show promise in preclinical or clinical studies for use in evaluating the phenotypic or biochemical characteristics of AD. Radiotracers for PET imaging neuroinflammation, metal ion association with Aβ-plaques, tau protein, cholinergic and cannabinoid receptors, and enzymes including glycogen-synthase kinase-3β and monoamine oxidase B amongst others, and their connection to AD are highlighted.
Collapse
Affiliation(s)
- Jason P Holland
- Division of Nuclear Medicine and Molecular Imaging, Massachusetts General Hospital, Department of Radiology, Harvard Medical School, 55 Fruit St., White 427, Boston, Massachusetts, 02114, USA; Life Sciences, Australian Nuclear Science and Technology Organisation, Kirrawee, New South Wales, 2232, Australia
| | | | | | | | | | | | | |
Collapse
|
50
|
Heng BC, Aubel D, Fussenegger M. An overview of the diverse roles of G-protein coupled receptors (GPCRs) in the pathophysiology of various human diseases. Biotechnol Adv 2013; 31:1676-94. [DOI: 10.1016/j.biotechadv.2013.08.017] [Citation(s) in RCA: 125] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2013] [Revised: 08/19/2013] [Accepted: 08/19/2013] [Indexed: 12/23/2022]
|