1
|
Lin J, Pang D, Li C, Ou R, Yu Y, Cui Y, Huang J, Shang H. Calcium channel blockers and Parkinson's disease: a systematic review and meta-analysis. Ther Adv Neurol Disord 2024; 17:17562864241252713. [PMID: 38770432 PMCID: PMC11104025 DOI: 10.1177/17562864241252713] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/22/2024] Open
Abstract
Background The calcium channel has been considered to have great potential as a drug target for neuroprotective therapy in Parkinson's disease (PD), but previous studies yielded inconsistent results. Objectives This study aimed to conduct a systematic review and meta-analysis to assess the relationship between using calcium channel blockers (CCBs) and the risk and progression of PD. Data sources and methods The terms such as 'Parkinson's disease', 'PD', 'calcium channel blockers', and 'CCB' were used to search the literature published before 1 May 2023 in English databases, including PubMed, Embase, and Cochrane Library, for studies on CCB and PD. Data analysis was performed using Review Manager 5.3 software. Results A total of 190 works of literature were preliminarily retrieved, and 177 works of literature were excluded by eliminating duplicates, reading abstracts, and reading full texts. A total of nine studies were finally included in the meta-analysis of the CCB and the risk of PD, and five studies were included in the systematic review of the CCB and the progression of PD. A total of 2,961,695 participants were included in the meta-analysis. The random-effects model was used for analysis due to significant heterogeneity. The main results of the meta-analysis showed that the use of CCB could reduce the risk of PD (relative risk 0.78, 95% confidence interval 0.62-0.99). Conclusion CCB use was associated with a significantly reduced risk of PD. Whether CCB use has a disease-modifying effect on PD needs further study. Registration PROSPERO: CRD42024508242.
Collapse
Affiliation(s)
- Junyu Lin
- Laboratory of Neurodegenerative Disorders, Department of Neurology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Dejiang Pang
- Laboratory of Neurodegenerative Disorders, Department of Neurology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Chunyu Li
- Laboratory of Neurodegenerative Disorders, Department of Neurology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Ruwei Ou
- Laboratory of Neurodegenerative Disorders, Department of Neurology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yujiao Yu
- Laboratory of Neurodegenerative Disorders, Department of Neurology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Yiyuan Cui
- Laboratory of Neurodegenerative Disorders, Department of Neurology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Jingxuan Huang
- Laboratory of Neurodegenerative Disorders, Department of Neurology, Rare Disease Center, West China Hospital, Sichuan University, Chengdu, Sichuan, China
| | - Huifang Shang
- Department of Neurology, West China Hospital, Sichuan University, Guoxue Road No. 37, Chengdu, Sichuan 610041, China
| |
Collapse
|
2
|
Wickline JL, Smith S, Shin R, Odfalk K, Sanchez J, Javors M, Ginsburg B, Hopp SC. L-type calcium channel antagonist isradipine age-dependently decreases plaque associated dystrophic neurites in 5XFAD mouse model. Neuropharmacology 2023; 227:109454. [PMID: 36740015 PMCID: PMC9987839 DOI: 10.1016/j.neuropharm.2023.109454] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2022] [Revised: 01/30/2023] [Accepted: 02/01/2023] [Indexed: 02/05/2023]
Abstract
Epidemiological studies suggest that L-type calcium channel (LTCC) antagonists may reduce the incidence of age-associated neurodegenerative diseases including Alzheimer's disease (AD). However, the neuroprotective mechanism of LTCC antagonists is unknown. Amyloid-β (Aβ) pathology disrupts intracellular calcium signaling, which regulates lysosomes and microglial responses. Neurons near Aβ plaques develop dystrophic neurites, which are abnormal swellings that accumulate lysosomes. Further, microglia accumulate around Aβ plaques and secrete inflammatory cytokines. We hypothesized that antagonism of LTCCs with isradipine would reduce Aβ plaque-associated dystrophic neurites and inflammatory microglia in the 5XFAD mouse model by restoring normal intracellular calcium regulation. To test this hypothesis, we treated 6- and 9-month-old 5XFAD mice with isradipine and tested behavior, examined Aβ plaques, microglia, and dystrophic neurites. We found that isradipine treatment age-dependently reduces dystrophic neurites and leads to trending decreases in Aβ but does not modulate plaque associated microglia regardless of age. Our findings provide insight into how antagonizing LTCCs alters specific cell types in the Aβ plaque environment, providing valuable information for potential treatment targets in future AD studies.
Collapse
Affiliation(s)
- Jessica L Wickline
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Sabrina Smith
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Riley Shin
- Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Kristian Odfalk
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| | - Jesse Sanchez
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Martin Javors
- Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Brett Ginsburg
- Department of Psychiatry and Behavioral Sciences, University of Texas Health Science Center at San Antonio, San Antonio, TX, USA
| | - Sarah C Hopp
- Glenn Biggs Institute for Alzheimer's and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA; Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA.
| |
Collapse
|
3
|
Suppression of Selective Voltage-Gated Calcium Channels Alleviates Neuronal Degeneration and Dysfunction through Glutathione S-Transferase-Mediated Oxidative Stress Resistance in a Caenorhabditis elegans Model of Alzheimer's Disease. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2022; 2022:8287633. [PMID: 36600949 PMCID: PMC9806690 DOI: 10.1155/2022/8287633] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/18/2022] [Accepted: 10/31/2022] [Indexed: 12/05/2022]
Abstract
Calcium homeostasis plays a vital role in protecting against Alzheimer's disease (AD). In this study, amyloid-β (Aβ)-induced C. elegans models of AD were used to elucidate the mechanisms underlying calcium homeostasis in AD. Calcium acetate increased the intracellular calcium content, exacerbated Aβ 1-42 aggregation, which is closely associated with oxidative stress, aggravated neuronal degeneration and dysfunction, and shortened the lifespan of the C. elegans models. Ethylene glycol tetraacetic acid (EGTA) and nimodipine were used to decrease the intracellular calcium content. Both EGTA and nimodipine showed remarkable inhibitory effects on Aβ 1-42 aggregations by increasing oxidative stress resistance. Moreover, both compounds significantly delayed the onset of Aβ-induced paralysis, rescued memory deficits, ameliorated behavioral dysfunction, decreased the vulnerability of two major (GABAergic and dopaminergic) neurons and synapses, and extended the lifespan of the C. elegans AD models. Furthermore, RNA sequencing of nimodipine-treated worms revealed numerous downstream differentially expressed genes related to calcium signaling. Nimodipine-induced inhibition of selective voltage-gated calcium channels was shown to activate other calcium channels of the plasma membrane (clhm-1) and endoplasmic reticulum (unc-68), in addition to sodium-calcium exchanger channels (ncx-1). These channels collaborated to activate downstream events to resist oxidative stress through glutathione S-transferase activity mediated by HPGD and skn-1, as verified by RNA interference. These results may be applied for the treatment of Alzheimer's disease.
Collapse
|
4
|
Thapak P, Khare P, Bishnoi M, Sharma SS. Neuroprotective Effect of 2-Aminoethoxydiphenyl Borate (2-APB) in Amyloid β-Induced Memory Dysfunction: A Mechanistic Study. Cell Mol Neurobiol 2022; 42:1211-1223. [PMID: 33219878 PMCID: PMC11441215 DOI: 10.1007/s10571-020-01012-z] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2020] [Accepted: 11/13/2020] [Indexed: 02/08/2023]
Abstract
β-Amyloid (Aβ) peptide is a characteristic feature of Alzheimer's disease (AD) and accumulation of Aβ is associated with loss of synaptic plasticity and neuronal cell death. Aggregation of Aβ initiates numerous molecular signalling pathways leading to oxidative stress, mitochondrial dysfunction as well as an imbalance of calcium ion influx homeostasis. Recently, it has been shown that transient receptor potential melastatin 2 (TRPM2), a non-selective calcium-permeable cation channel has been postulated to play a vital role in the neuronal death, indicating the potential of TRPM2 inhibition in CNS disease. In this study, neuroprotective potential of 2-aminoethoxydiphenyl borate (2-APB), a broad-spectrum calcium channels blocker was investigated in Aβ-induced memory deficits in rats. In addition, effect of 2-APB on TRPM2 channels gene and protein expressions and also on calcium and memory related proteins was investigated in the hippocampus. Intracerebroventricular (I.C.V.) administration of Aβ (Aβ25-35, 10 μg) markedly induced cognitive impairment and upregulation of mRNA and protein expression of TRPM2 in the hippocampus. In addition, AChE activity was also increased in the cortex of the Aβ administered animals. Three-week treatment with 2-APB led to the down-regulation of TRPM2 mRNA and protein expression in the hippocampus and also improved the cognitive functions which was evident from the behavioral parameters. Moreover, 2-APB treatment also increased the calcium and memory associated proteins namely p-CaMKII, p-GSK-3β, p-CREB and PSD-95 in the hippocampus and reduced the mRNA level of calcium buffering proteins and calcineurin A (PPP3CA) in the hippocampus. Furthermore, 2-APB treatment significantly reduced the AChE activity in the cortex. Thus, our findings suggest the neuroprotective effect of 2-APB in Aβ-induced cognitive impairment.
Collapse
Affiliation(s)
- Pavan Thapak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Pragyanshu Khare
- National Agri-Food Biotechnology Institute (NABI), S.A.S. Nagar, Mohali, Punjab, India
| | - Mahendra Bishnoi
- National Agri-Food Biotechnology Institute (NABI), S.A.S. Nagar, Mohali, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), S.A.S. Nagar, Mohali, Punjab, 160062, India.
| |
Collapse
|
5
|
Hohmann U, Ghadban C, Hohmann T, Kleine J, Schmidt M, Scheller C, Strauss C, Dehghani F. Nimodipine Exerts Time-Dependent Neuroprotective Effect after Excitotoxical Damage in Organotypic Slice Cultures. Int J Mol Sci 2022; 23:ijms23063331. [PMID: 35328753 PMCID: PMC8954806 DOI: 10.3390/ijms23063331] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Revised: 03/14/2022] [Accepted: 03/16/2022] [Indexed: 02/01/2023] Open
Abstract
During injuries in the central nervous system, intrinsic protective processes become activated. However, cellular reactions, especially those of glia cells, are frequently unsatisfactory, and further exogenous protective mechanisms are necessary. Nimodipine, a lipophilic L-type calcium channel blocking agent is clinically used in the treatment of aneurysmal subarachnoid haemorrhage with neuroprotective effects in different models. Direct effects of nimodipine on neurons amongst others were observed in the hippocampus as well as its influence on both microglia and astrocytes. Earlier studies proposed that nimodipine protective actions occur not only via calcium channel-mediated vasodilatation but also via further time-dependent mechanisms. In this study, the effect of nimodipine application was investigated in different time frames on neuronal damage in excitotoxically lesioned organotypic hippocampal slice cultures. Nimodipine, but not nifedipine if pre-incubated for 4 h or co-applied with NMDA, was protective, indicating time dependency. Since blood vessels play no significant role in our model, intrinsic brain cell-dependent mechanisms seems to strongly be involved. We also examined the effect of nimodipine and nifedipine on microglia survival. Nimodipine seem to be a promising agent to reduce secondary damage and reduce excitotoxic damage.
Collapse
Affiliation(s)
- Urszula Hohmann
- Medical Faculty, Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (U.H.); (C.G.); (T.H.); (J.K.); (M.S.)
| | - Chalid Ghadban
- Medical Faculty, Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (U.H.); (C.G.); (T.H.); (J.K.); (M.S.)
| | - Tim Hohmann
- Medical Faculty, Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (U.H.); (C.G.); (T.H.); (J.K.); (M.S.)
| | - Joshua Kleine
- Medical Faculty, Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (U.H.); (C.G.); (T.H.); (J.K.); (M.S.)
| | - Miriam Schmidt
- Medical Faculty, Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (U.H.); (C.G.); (T.H.); (J.K.); (M.S.)
| | - Christian Scheller
- Department of Neurosurgery, Medical Faculty, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (C.S.); (C.S.)
| | - Christian Strauss
- Department of Neurosurgery, Medical Faculty, Martin Luther University Halle-Wittenberg, 06120 Halle (Saale), Germany; (C.S.); (C.S.)
| | - Faramarz Dehghani
- Medical Faculty, Institute of Anatomy and Cell Biology, Martin Luther University Halle-Wittenberg, 06112 Halle (Saale), Germany; (U.H.); (C.G.); (T.H.); (J.K.); (M.S.)
- Correspondence: ; Tel.: +49-3455571707
| |
Collapse
|
6
|
Thapak P, Bishnoi M, Sharma SS. Tranilast, a Transient Receptor Potential Vanilloid 2 Channel (TRPV2) Inhibitor Attenuates Amyloid β-Induced Cognitive Impairment: Possible Mechanisms. Neuromolecular Med 2021; 24:183-194. [PMID: 34231190 DOI: 10.1007/s12017-021-08675-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2021] [Accepted: 06/24/2021] [Indexed: 02/08/2023]
Abstract
Alzheimer's disease (AD) is associated with the accumulation of β-amyloid and leads to cognitive impairment. Numerous studies have established that neuronal calcium homeostasis is perturbed in AD. Recently, transient receptor potential vanilloid 2 (TRPV2) channels, a non-selective calcium-permeable channel, have been investigated in several diseases. However, the role of the TRPV2 channel has not been investigated in AD yet. In this study, intracerebroventricular administration of β-amyloid (10 μg) to Sprague Dawley rats resulted in cognitive impairment which was evident from the assessment of cognitive tests. Also, TRPV2 mRNA and protein expression were found to be upregulated, while the expression of Ca2+/calmodulin-dependent protein kinase II (p-CaMKII-Thr-286), glycogen synthase kinase 3β (p-GSK-3β-Ser-9), cAMP response element-binding protein (p-CREB-Ser-133), and postsynaptic density protein 95 (PSD-95) were downregulated in the hippocampus of β-amyloid-treated animals. Even, β-amyloid-treated animals showed upregulation of mRNA level of calcium buffering proteins (parvalbumin and calsequestrin) and calcineurin A (PPP3CA) in the hippocampus. Acetylcholinesterase activity was also increased in the cortex of β-amyloid-treated animals. Three-week treatment with tranilast showed improvement in the cognitive parameters which was associated with a decrease in TRPV2 expression and AChE activity. Additionally, an increase in the protein expression of p-CaMKII, p-GSK-3β, p-CREB and PSD-95 in the hippocampus was found. Downregulation in the mRNA level of calcium buffering proteins (parvalbumin and calsequestrin) and calcineurin A in the hippocampus was also seen. These results reveal the importance of TRPV2 channels in the β-amyloid-induced cognitive deficits and suggest TRPV2 as a potential target for AD.
Collapse
Affiliation(s)
- Pavan Thapak
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, Punjab, 160062, India
| | - Mahendra Bishnoi
- National Agri-Food Biotechnology Institute, S.A.S. Nagar, Punjab, India
| | - Shyam Sunder Sharma
- Department of Pharmacology and Toxicology, National Institute of Pharmaceutical Education and Research (NIPER), Sector 67, S.A.S. Nagar, Mohali, Punjab, 160062, India.
| |
Collapse
|
7
|
Yakupova EI, Bobyleva LG, Shumeyko SA, Vikhlyantsev IM, Bobylev AG. Amyloids: The History of Toxicity and Functionality. BIOLOGY 2021; 10:biology10050394. [PMID: 34062910 PMCID: PMC8147320 DOI: 10.3390/biology10050394] [Citation(s) in RCA: 18] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/06/2021] [Revised: 04/26/2021] [Accepted: 04/29/2021] [Indexed: 12/15/2022]
Abstract
Proteins can perform their specific function due to their molecular structure. Partial or complete unfolding of the polypeptide chain may lead to the misfolding and aggregation of proteins in turn, resulting in the formation of different structures such as amyloid aggregates. Amyloids are rigid protein aggregates with the cross-β structure, resistant to most solvents and proteases. Because of their resistance to proteolysis, amyloid aggregates formed in the organism accumulate in tissues, promoting the development of various diseases called amyloidosis, for instance Alzheimer's diseases (AD). According to the main hypothesis, it is considered that the cause of AD is the formation and accumulation of amyloid plaques of Aβ. That is why Aβ-amyloid is the most studied representative of amyloids. Therefore, in this review, special attention is paid to the history of Aβ-amyloid toxicity. We note the main problems with anti-amyloid therapy and write about new views on amyloids that can play positive roles in the different organisms including humans.
Collapse
Affiliation(s)
- Elmira I. Yakupova
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (L.G.B.); (S.A.S.); (I.M.V.); (A.G.B.)
- A. N. Belozersky Institute of Physico-Chemical Biology, Lomonosov Moscow State University, 119991 Moscow, Russia
- Correspondence: ; Tel.: +7-(985)687-77-27
| | - Liya G. Bobyleva
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (L.G.B.); (S.A.S.); (I.M.V.); (A.G.B.)
| | - Sergey A. Shumeyko
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (L.G.B.); (S.A.S.); (I.M.V.); (A.G.B.)
| | - Ivan M. Vikhlyantsev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (L.G.B.); (S.A.S.); (I.M.V.); (A.G.B.)
| | - Alexander G. Bobylev
- Institute of Theoretical and Experimental Biophysics, Russian Academy of Sciences, Pushchino, 142290 Moscow, Russia; (L.G.B.); (S.A.S.); (I.M.V.); (A.G.B.)
| |
Collapse
|
8
|
Duan X, Li Y, Xu F, Ding H. Study on the neuroprotective effects of Genistein on Alzheimer's disease. Brain Behav 2021; 11:e02100. [PMID: 33704934 PMCID: PMC8119804 DOI: 10.1002/brb3.2100] [Citation(s) in RCA: 40] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Revised: 01/04/2021] [Accepted: 02/23/2021] [Indexed: 01/14/2023] Open
Abstract
Along with the aging of the world population, the incidence rate of Alzheimer's disease (AD) has been increasing. At present, AD has become one of the most serious problems faced by modern medicine. Studies have shown that estrogen has a positive effect on AD, but estrogen has the side effect of leading to tumors. Recent in vivo studies have shown that genistein, one of the selective estrogen receptor modulators (SERMs), can improve brain function through the blood-brain barrier (BBB), antagonize the toxicity of amyloid β-protein (Aβ), that is, to inhibit neurotoxicity due to aggregation of beta amyloid protein, and have neuroprotective effects. In addition, the use of Gen can avoid the risk of endometrial cancer and breast cancer caused by estrogen therapy while exerting an estrogen-like effect, which has some potential for the delay and treatment of AD.
Collapse
Affiliation(s)
- Xiaoying Duan
- Department of Acupuncture and Moxibustion, the Second Hospital of Jilin University, Changchun, China
| | - Yanshuang Li
- Department of Acupuncture and Moxibustion, the Second Hospital of Jilin University, Changchun, China
| | - Fei Xu
- Department of Acupuncture and Moxibustion, the Second Hospital of Jilin University, Changchun, China
| | - Hong Ding
- Department of Traditional Chinese Medicine, the Second Hospital of Jilin University, Changchun, China
| |
Collapse
|
9
|
Hopp SC. Targeting microglia L-type voltage-dependent calcium channels for the treatment of central nervous system disorders. J Neurosci Res 2021; 99:141-162. [PMID: 31997405 PMCID: PMC9394523 DOI: 10.1002/jnr.24585] [Citation(s) in RCA: 26] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2019] [Revised: 01/03/2020] [Accepted: 01/08/2020] [Indexed: 12/14/2022]
Abstract
Calcium (Ca2+ ) is a ubiquitous mediator of a multitude of cellular functions in the central nervous system (CNS). Intracellular Ca2+ is tightly regulated by cells, including entry via plasma membrane Ca2+ permeable channels. Of specific interest for this review are L-type voltage-dependent Ca2+ channels (L-VDCCs), due to their pleiotropic role in several CNS disorders. Currently, there are numerous approved drugs that target L-VDCCs, including dihydropyridines. These drugs are safe and effective for the treatment of humans with cardiovascular disease and may also confer neuroprotection. Here, we review the potential of L-VDCCs as a target for the treatment of CNS disorders with a focus on microglia L-VDCCs. Microglia, the resident immune cells of the brain, have attracted recent attention for their emerging inflammatory role in several CNS diseases. Intracellular Ca2+ regulates microglia transition from a resting quiescent state to an "activated" immune-effector state and is thus a valuable target for manipulation of microglia phenotype. We will review the literature on L-VDCC expression and function in the CNS and on microglia in vitro and in vivo and explore the therapeutic landscape of L-VDCC-targeting agents at present and future challenges in the context of Alzheimer's disease, Parkinson's disease, Huntington's disease, neuropsychiatric diseases, and other CNS disorders.
Collapse
Affiliation(s)
- Sarah C. Hopp
- Glenn Biggs Institute for Alzheimer’s and Neurodegenerative Diseases, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
- Department of Pharmacology, University of Texas Health Science Center San Antonio, San Antonio, TX, USA
| |
Collapse
|
10
|
Oh SJ, Lee HJ, Jeong YJ, Nam KR, Kang KJ, Han SJ, Lee KC, Lee YJ, Choi JY. Evaluation of the neuroprotective effect of taurine in Alzheimer's disease using functional molecular imaging. Sci Rep 2020; 10:15551. [PMID: 32968166 PMCID: PMC7511343 DOI: 10.1038/s41598-020-72755-4] [Citation(s) in RCA: 28] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/10/2020] [Accepted: 09/04/2020] [Indexed: 01/22/2023] Open
Abstract
Alzheimer's disease (AD) is a chronic neurodegenerative disorder and the leading cause of dementia, but therapeutic treatment options are limited. Taurine has been reported to have neuroprotective properties against dementia, including AD. The present study aimed to investigate the treatment effect of taurine in AD mice by functional molecular imaging. To elucidate glutamate alterations by taurine, taurine was administered to 5xFAD transgenic mice from 2 months of age, known to apear amyloid deposition. Then, we performed glutamate positron emission tomography (PET) imaging studies for three groups (wild-type, AD, and taurine-treated AD, n = 5 in each group). As a result, brain uptake in the taurine-treated AD group was 31-40% higher than that in the AD group (cortex: 40%, p < 0.05; striatum: 32%, p < 0.01; hippocampus: 36%, p < 0.01; thalamus: 31%, p > 0.05) and 3-14% lower than that in the WT group (cortex: 10%, p > 0.05; striatum: 15%, p > 0.05; hippocampus: 14%, p > 0.05; thalamus: 3%, p > 0.05). However, we did not observe differences in Aβ pathology between the taurine-treated AD and AD groups in immunohistochemistry experiments. Our results reveal that although taurine treatment did not completely recover the glutamate system, it significantly increased metabolic glutamate receptor type 5 brain uptake. Therefore, taurine has therapeutic potential against AD.
Collapse
Affiliation(s)
- Se Jong Oh
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Korea
| | - Hae-June Lee
- Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Ye Ji Jeong
- Division of Radiation Effects, Korea Institute of Radiological and Medical Sciences, Seoul, Korea
| | - Kyung Rok Nam
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Korea
| | - Kyung Jun Kang
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Korea
| | - Sang Jin Han
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Korea
| | - Kyo Chul Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Korea
| | - Yong Jin Lee
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Korea
| | - Jae Yong Choi
- Division of Applied RI, Korea Institute of Radiological and Medical Sciences, 75 Nowon-ro, Nowon-gu, Seoul, 01812, Korea.
| |
Collapse
|
11
|
Lourdes A. Vega Rasgado, Urbieta AT, Medina Jiménez JM. Influence of Mitochondrial ATP-Sensitive Potassium Channels on Toxic Effect of Amyloid-β 25–35. NEUROCHEM J+ 2020. [DOI: 10.1134/s181971242001016x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
12
|
Carlson AP, Hänggi D, Macdonald RL, Shuttleworth CW. Nimodipine Reappraised: An Old Drug With a Future. Curr Neuropharmacol 2020; 18:65-82. [PMID: 31560289 PMCID: PMC7327937 DOI: 10.2174/1570159x17666190927113021] [Citation(s) in RCA: 87] [Impact Index Per Article: 17.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2019] [Revised: 07/02/2019] [Accepted: 09/25/2019] [Indexed: 12/21/2022] Open
Abstract
Nimodipine is a dihydropyridine calcium channel antagonist that blocks the flux of extracellular calcium through L-type, voltage-gated calcium channels. While nimodipine is FDAapproved for the prevention and treatment of neurological deficits in patients with aneurysmal subarachnoid hemorrhage (aSAH), it affects myriad cell types throughout the body, and thus, likely has more complex mechanisms of action than simple inhibition of cerebral vasoconstriction. Newer understanding of the pathophysiology of delayed ischemic injury after a variety of acute neurologic injuries including aSAH, traumatic brain injury (TBI) and ischemic stroke, coupled with advances in the drug delivery method for nimodipine, have reignited interest in refining its potential therapeutic use. In this context, this review seeks to establish a firm understanding of current data on nimodipine's role in the mechanisms of delayed injury in aSAH, TBI, and ischemic stroke, and assess the extensive clinical data evaluating its use in these conditions. In addition, we will review pivotal trials using locally administered, sustained release nimodipine and discuss why such an approach has evaded demonstration of efficacy, while seemingly having the potential to significantly improve clinical care.
Collapse
Affiliation(s)
- Andrew P. Carlson
- Department of Neurosurgery, University of New Mexico School of Medicine, Albuquerque, NM, USA
| | - Daniel Hänggi
- Department of Neurosurgery, University of Dusseldorf Hospital, Heinrich-Heine-Universität, Düsseldorf, Germany
| | - Robert L. Macdonald
- University of California San Francisco Fresno Department of Neurosurgery and University Neurosciences Institute and Division of Neurosurgery, Department of Surgery, University of Toronto, Canada
| | - Claude W. Shuttleworth
- Department of Neuroscience University of New Mexico School of Medicine, Albuquerque, NM, USA
| |
Collapse
|
13
|
Lebouvier T, Chen Y, Duriez P, Pasquier F, Bordet R. Antihypertensive agents in Alzheimer's disease: beyond vascular protection. Expert Rev Neurother 2019; 20:175-187. [PMID: 31869274 DOI: 10.1080/14737175.2020.1708195] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Introduction: Midlife hypertension has been consistently linked with increased risk of cognitive decline and Alzheimer's disease (AD). Observational studies and randomized trials show that the use of antihypertensive therapy is associated with a lesser incidence or prevalence of cognitive impairment and dementia. However, whether antihypertensive agents specifically target the pathological process of AD remains elusive.Areas covered: This review of literature provides an update on the clinical and preclinical arguments supporting anti-AD properties of antihypertensive drugs. The authors focused on validated all classes of antihypertensive treatments such as angiotensin-converting enzyme inhibitors (ACEi), angiotensin receptor blockers (ARB), calcium channel blockers (CCB), β-blockers, diuretics, neprilysin inhibitors, and other agents. Three main mechanisms can be advocated: action on the concurrent vascular pathology, action on the vascular component of Alzheimer's pathophysiology, and action on nonvascular targets.Expert opinion: In 2019, while there is no doubt that hypertension should be treated in primary prevention of vascular disease and in secondary prevention of stroke and mixed dementia, the place of antihypertensive agents in the secondary prevention of 'pure' AD remains an outstanding question.
Collapse
Affiliation(s)
- Thibaud Lebouvier
- Inserm URM_S1172, University of Lille, Lille, France.,DISTALZ, University of Lille, Lille, France
| | - Yaohua Chen
- DISTALZ, University of Lille, Lille, France.,Inserm, CHU Lille, University of Lille, Lille, France
| | | | - Florence Pasquier
- DISTALZ, University of Lille, Lille, France.,Inserm, CHU Lille, University of Lille, Lille, France
| | - Régis Bordet
- Inserm, CHU Lille, University of Lille, Lille, France
| |
Collapse
|
14
|
Amyloid-Beta Modulates Low-Threshold Activated Voltage-Gated L-Type Calcium Channels of Arcuate Neuropeptide Y Neurons Leading to Calcium Dysregulation and Hypothalamic Dysfunction. J Neurosci 2019; 39:8816-8825. [PMID: 31537707 DOI: 10.1523/jneurosci.0617-19.2019] [Citation(s) in RCA: 33] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2019] [Revised: 07/17/2019] [Accepted: 09/11/2019] [Indexed: 11/21/2022] Open
Abstract
Weight loss is an early manifestation of Alzheimer's disease that can precede the cognitive decline, raising the possibility that amyloid-β (Aβ) disrupts hypothalamic neurons critical for the regulation of body weight. We previously reported that, in young transgenic mice overexpressing mutated amyloid precursor protein (Tg2576), Aβ causes dysfunction in neuropeptide Y (NPY)-expressing hypothalamic arcuate neurons before plaque formation. In this study, we examined whether Aβ causes arcuate NPY neuronal dysfunction by disrupting intracellular Ca2+ homeostasis. Here, we found that the L-type Ca2+ channel blocker nimodipine could hyperpolarize the membrane potential, decrease the spontaneous activity, and reduce the intracellular Ca2+ levels in arcuate NPY neurons from Tg2576 brain slices. In these neurons, there was a shift from high to low voltage-threshold activated L-type Ca2+ currents, resulting in increased Ca2+ influx closer to the resting membrane potential, an effect recapitulated by Aβ1-42 and reversed by nimodipine. These low voltage-threshold activated L-type Ca2+ currents were dependent in part on calcium/calmodulin-dependent protein kinase II and IP3 pathways. Furthermore, the effects on intracellular Ca2+ signaling by both a positive (ghrelin) and negative (leptin) modulator were blunted in these neurons. Nimodipine pretreatment restored the response to ghrelin-mediated feeding in young (3-5 months), but not older (10 months), female Tg2576 mice, suggesting that intracellular Ca2+ dysregulation is only reversible early in Aβ pathology. Collectively, these findings provide evidence for a key role for low-threshold activated voltage gated L-type Ca2+ channels in Aβ-mediated neuronal dysfunction and in the regulation of body weight.SIGNIFICANCE STATEMENT Weight loss is one of the earliest manifestations of Alzheimer's disease (AD), but the underlying cellular mechanisms remain unknown. Disruption of intracellular Ca2+ homeostasis by amyloid-β is hypothesized to be critical for the early neuronal dysfunction driving AD pathogenesis. Here, we demonstrate that amyloid-β causes a shift from high to low voltage-threshold activated L-type Ca2+ currents in arcuate neuropeptide Y neurons. This leads to increased Ca2+ influx closer to the resting membrane potential, resulting in intracellular Ca2+ dyshomeostasis and neuronal dysfunction, an effect reversible by the L-type Ca2+ channel blocker nimodipine early in amyloid-β pathology. These findings highlight a novel mechanism of amyloid-β-mediated neuronal dysfunction through L-type Ca2+ channels and the importance of these channels in the regulation of body weight.
Collapse
|
15
|
Luo H, Han L, Xu J. Apelin/APJ system: A novel promising target for neurodegenerative diseases. J Cell Physiol 2019; 235:638-657. [DOI: 10.1002/jcp.29001] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2019] [Accepted: 06/06/2019] [Indexed: 12/17/2022]
Affiliation(s)
- Huaiqing Luo
- Department of Physiology Changsha Medical University Changsha Hunan China
- Department of Physiology, School of Basic Medical Science Central South University Changsha Hunan China
| | - Li Han
- Department of Physiology Changsha Medical University Changsha Hunan China
| | - Jin Xu
- School of Pharmaceutical Sciences Changsha Medical University Changsha Hunan China
| |
Collapse
|
16
|
Ge X, Sun Y, Ding F. Structures and dynamics of β-barrel oligomer intermediates of amyloid-beta16-22 aggregation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2018; 1860:1687-1697. [PMID: 29550287 DOI: 10.1016/j.bbamem.2018.03.011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/31/2017] [Revised: 03/06/2018] [Accepted: 03/07/2018] [Indexed: 01/13/2023]
Abstract
Accumulating evidence suggests that soluble oligomers are more toxic than final fibrils of amyloid aggregations. Among the mixture of inter-converting intermediates with continuous distribution of sizes and secondary structures, oligomers in the β-barrel conformation - a common class of protein folds with a closed β-sheet - have been postulated as the toxic species with well-defined three-dimensional structures to perform pathological functions. A common mechanism for amyloid toxicity, therefore, implies that all amyloid peptides should be able to form β-barrel oligomers as the aggregation intermediates. Here, we applied all-atom discrete molecular dynamics (DMD) simulations to evaluate the formation of β-barrel oligomers and characterize their structures and dynamics in the aggregation of a seven-residue amyloid peptide, corresponding to the amyloid core of amyloid-β with a sequence of 16KLVFFAE22 (Aβ16-22). We carried out aggregation simulations with various numbers of peptides to study the size dependence of aggregation dynamics and assembly structures. Consistent with previous computational studies, we observed the formation of β-barrel oligomers in all-atom DMD simulations. Using a network-based approach to automatically identify β-barrel conformations, we systematically characterized β-barrels of various sizes. Our simulations revealed the conformational inter-conversion between β-barrels and double-layer β-sheets due to increased structural strains upon forming a closed β-barrel while maximizing backbone hydrogen bonds. The potential of mean force analysis further characterized the free energy barriers between these two states. The obtained structural and dynamic insights of β-barrel oligomers may help better understand the molecular mechanism of oligomer toxicities and design novel therapeutics targeting the toxic β-barrel oligomers. This article is part of a Special Issue entitled: Protein Aggregation and Misfolding at the Cell Membrane Interface edited by Ayyalusamy Ramamoorthy.
Collapse
Affiliation(s)
- Xinwei Ge
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Yunxiang Sun
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States
| | - Feng Ding
- Department of Physics and Astronomy, Clemson University, Clemson, SC 29634, United States.
| |
Collapse
|
17
|
Ingwersen J, De Santi L, Wingerath B, Graf J, Koop B, Schneider R, Hecker C, Schröter F, Bayer M, Engelke AD, Dietrich M, Albrecht P, Hartung HP, Annunziata P, Aktas O, Prozorovski T. Nimodipine confers clinical improvement in two models of experimental autoimmune encephalomyelitis. J Neurochem 2018; 146:86-98. [PMID: 29473171 DOI: 10.1111/jnc.14324] [Citation(s) in RCA: 22] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Revised: 02/07/2018] [Accepted: 02/08/2018] [Indexed: 12/31/2022]
Abstract
Multiple sclerosis is characterised by inflammatory neurodegeneration, with axonal injury and neuronal cell death occurring in parallel to demyelination. Regarding the molecular mechanisms responsible for demyelination and axonopathy, energy failure, aberrant expression of ion channels and excitotoxicity have been suggested to lead to Ca2+ overload and subsequent activation of calcium-dependent damage pathways. Thus, the inhibition of Ca2+ influx by pharmacological modulation of Ca2+ channels may represent a novel neuroprotective strategy in the treatment of secondary axonopathy. We therefore investigated the effects of the L-type voltage-gated calcium channel blocker nimodipine in two different models of mouse experimental autoimmune encephalomyelitis (EAE), an established experimental paradigm for multiple sclerosis. We show that preventive application of nimodipine (10 mg/kg per day) starting on the day of induction had ameliorating effects on EAE in SJL/J mice immunised with encephalitic myelin peptide PLP139-151 , specifically in late-stage disease. Furthermore, supporting these data, administration of nimodipine to MOG35-55 -immunised C57BL/6 mice starting at the peak of pre-established disease, also led to a significant decrease in disease score, indicating a protective effect on secondary CNS damage. Histological analysis confirmed that nimodipine attenuated demyelination, axonal loss and pathological axonal β-amyloid precursor protein accumulation in the cerebellum and spinal cord in the chronic phase of disease. Of note, we observed no effects of nimodipine on the peripheral immune response in EAE mice with regard to distribution, antigen-specific proliferation or activation patterns of lymphocytes. Taken together, our data suggest a CNS-specific effect of L-type voltage-gated calcium channel blockade to inflammation-induced neurodegeneration.
Collapse
Affiliation(s)
- Jens Ingwersen
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Lorenzo De Santi
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Britta Wingerath
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Jonas Graf
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Barbara Koop
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Reiner Schneider
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Christina Hecker
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Friederike Schröter
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Mary Bayer
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Anna Dorothee Engelke
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Michael Dietrich
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Philipp Albrecht
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Hans-Peter Hartung
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Pasquale Annunziata
- Department of Medicine, Surgery and Neuroscience, University of Siena, Siena, Italy
| | - Orhan Aktas
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| | - Tim Prozorovski
- Department of Neurology, Medical Faculty, Heinrich-Heine-University, Düsseldorf, Germany
| |
Collapse
|
18
|
Martin-de-Saavedra MD, Navarro E, Moreno-Ortega AJ, Cunha MP, Buendia I, Hernansanz-Agustín P, León R, Cano-Abad MF, Martínez-Ruiz A, Martínez-Murillo R, Duchen MR, López MG. The APPswe/PS1A246E mutations in an astrocytic cell line leads to increased vulnerability to oxygen and glucose deprivation, Ca2+
dysregulation, and mitochondrial abnormalities. J Neurochem 2018; 145:170-182. [DOI: 10.1111/jnc.14293] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2017] [Revised: 11/20/2017] [Accepted: 12/08/2017] [Indexed: 01/03/2023]
Affiliation(s)
- María Dolores Martin-de-Saavedra
- Instituto Teófilo Hernando; Universidad Autónoma de Madrid; Madrid Spain
- Departamento de Farmacología y Terapéutica; Facultad de Medicina; Instituto de Investigación Sanitaria Princesa (IIS-IP); Universidad Autónoma de Madrid; Madrid Spain
- Department of Physiology; Northwestern University Feinberg School of Medicine; Chicago Illinois USA
| | - Elisa Navarro
- Instituto Teófilo Hernando; Universidad Autónoma de Madrid; Madrid Spain
- Departamento de Farmacología y Terapéutica; Facultad de Medicina; Instituto de Investigación Sanitaria Princesa (IIS-IP); Universidad Autónoma de Madrid; Madrid Spain
| | - Ana J. Moreno-Ortega
- Instituto Teófilo Hernando; Universidad Autónoma de Madrid; Madrid Spain
- Departamento de Farmacología y Terapéutica; Facultad de Medicina; Instituto de Investigación Sanitaria Princesa (IIS-IP); Universidad Autónoma de Madrid; Madrid Spain
- Servicio de Farmacología Clínica; Instituto de Investigación Sanitaria Princesa (IIS-IP); Hospital Universitario de la Princesa; Madrid Spain
| | - Mauricio P. Cunha
- Departamento de Bioquímica; Universidade Federal de Santa Catarina; Florianópolis Brazil
| | - Izaskun Buendia
- Instituto Teófilo Hernando; Universidad Autónoma de Madrid; Madrid Spain
- Departamento de Farmacología y Terapéutica; Facultad de Medicina; Instituto de Investigación Sanitaria Princesa (IIS-IP); Universidad Autónoma de Madrid; Madrid Spain
| | - Pablo Hernansanz-Agustín
- Servicio de Inmunología; Instituto de Investigación Sanitaria Princesa (IIS-IP); Hospital Universitario de la Princesa; Madrid Spain
- Departamento de Bioquímica; Facultad de Medicina; Universidad Autónoma de Madrid (UAM) and Instituto de Investigaciones Biomédicas Alberto Sols; Madrid Spain
| | - Rafael León
- Instituto Teófilo Hernando; Universidad Autónoma de Madrid; Madrid Spain
- Departamento de Farmacología y Terapéutica; Facultad de Medicina; Instituto de Investigación Sanitaria Princesa (IIS-IP); Universidad Autónoma de Madrid; Madrid Spain
- Servicio de Farmacología Clínica; Instituto de Investigación Sanitaria Princesa (IIS-IP); Hospital Universitario de la Princesa; Madrid Spain
| | - María F. Cano-Abad
- Instituto Teófilo Hernando; Universidad Autónoma de Madrid; Madrid Spain
- Departamento de Farmacología y Terapéutica; Facultad de Medicina; Instituto de Investigación Sanitaria Princesa (IIS-IP); Universidad Autónoma de Madrid; Madrid Spain
- Servicio de Farmacología Clínica; Instituto de Investigación Sanitaria Princesa (IIS-IP); Hospital Universitario de la Princesa; Madrid Spain
| | - Antonio Martínez-Ruiz
- Servicio de Inmunología; Instituto de Investigación Sanitaria Princesa (IIS-IP); Hospital Universitario de la Princesa; Madrid Spain
- Centro de Investigación Biomédica en Red de Enfermedades Cardiovasculares (CIBERCV); Madrid Spain
| | | | - Michael R. Duchen
- Department of Cell and Developmental Biology; University College London; London UK
| | - Manuela G. López
- Instituto Teófilo Hernando; Universidad Autónoma de Madrid; Madrid Spain
- Departamento de Farmacología y Terapéutica; Facultad de Medicina; Instituto de Investigación Sanitaria Princesa (IIS-IP); Universidad Autónoma de Madrid; Madrid Spain
| |
Collapse
|
19
|
Schampel A, Kuerten S. Danger: High Voltage-The Role of Voltage-Gated Calcium Channels in Central Nervous System Pathology. Cells 2017; 6:E43. [PMID: 29140302 PMCID: PMC5755501 DOI: 10.3390/cells6040043] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/14/2017] [Revised: 11/10/2017] [Accepted: 11/13/2017] [Indexed: 11/17/2022] Open
Abstract
Voltage-gated calcium channels (VGCCs) are widely distributed within the central nervous system (CNS) and presumed to play an important role in the pathophysiology of a broad spectrum of CNS disorders including Alzheimer's and Parkinson's disease as well as multiple sclerosis. Several calcium channel blockers have been in clinical practice for many years so that their toxicity and side effects are well studied. However, these drugs are primarily used for the treatment of cardiovascular diseases and most if not all effects on brain functions are secondary to peripheral effects on blood pressure and circulation. While the use of calcium channel antagonists for the treatment of CNS diseases therefore still heavily depends on the development of novel strategies to specifically target different channels and channel subunits, this review is meant to provide an impulse to further emphasize the importance of future research towards this goal.
Collapse
Affiliation(s)
- Andrea Schampel
- Institute of Anatomy and Cell Biology, University of Würzburg, Würzburg 97070, Germany.
| | - Stefanie Kuerten
- Institute of Anatomy and Cell Biology, Friedrich-Alexander University Erlangen-Nürnberg, Erlangen 91054, Germany.
| |
Collapse
|
20
|
Sainaghi PP, Bellan M, Lombino F, Alciato F, Carecchio M, Galimberti D, Fenoglio C, Scarpini E, Cantello R, Pirisi M, Comi C. Growth Arrest Specific 6 Concentration is Increased in the Cerebrospinal Fluid of Patients with Alzheimer's Disease. J Alzheimers Dis 2017; 55:59-65. [PMID: 27636849 DOI: 10.3233/jad-160599] [Citation(s) in RCA: 39] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
Growth arrest specific 6 (Gas6) has neurotrophic and neuroinflammatory functions, and may play a role in Alzheimer's disease (AD). In keeping with this hypothesis, we observed that cerebrospinal fluid (CSF) Gas6 is increased in AD patients compared to controls (63 versus 67 subjects; median value 13.3 versus 9.1 ng/ml; p < 0.0001). Thereafter, we assessed whether CSF Gas6 concentration was correlated to the following parameters: disease duration, MMSE score two years after clinical diagnosis, AD CSF biomarkers, and years of formal schooling. We detected an inverse correlation between CSF Gas6 levels at diagnosis and both disease duration (p < 0.0001) and decrease in the MMSE score two years later (p < 0.0001). Conversely, we found no correlation between CSF Gas6 and both AD biomarkers and years of formal schooling. In conclusion, our results suggest that upregulation of CSF Gas6 may be part of a defensive response aimed at counteracting AD progression.
Collapse
Affiliation(s)
- Pier Paolo Sainaghi
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
- Immuno-Rheumatology Unit, Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara, Italy
| | - Mattia Bellan
- Immuno-Rheumatology Unit, Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara, Italy
- Department of Translational Medicine, Internal Medicine, University of Piemonte Orientale, Novara, Italy
| | - Franco Lombino
- Department Molecular Neurogenetics, Center for Molecular Neurobiology (ZMNH), University of Hamburg Medical Center (UKE), Hamburg, Germany
| | - Federica Alciato
- Department of Translational Medicine, Internal Medicine, University of Piemonte Orientale, Novara, Italy
| | - Miryam Carecchio
- Department of Translational Medicine, Neurology Unit, University of Piemonte Orientale, Novara, Italy
| | - Daniela Galimberti
- Department of Pathophysiology and Transplantation, Neurology Unit, University of Milan, Fondazione Ca' Granda, IRCCS Ospedale Policlinico, Milan, Italy
| | - Chiara Fenoglio
- Department of Pathophysiology and Transplantation, Neurology Unit, University of Milan, Fondazione Ca' Granda, IRCCS Ospedale Policlinico, Milan, Italy
| | - Elio Scarpini
- Department of Pathophysiology and Transplantation, Neurology Unit, University of Milan, Fondazione Ca' Granda, IRCCS Ospedale Policlinico, Milan, Italy
| | - Roberto Cantello
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
- Department of Translational Medicine, Neurology Unit, University of Piemonte Orientale, Novara, Italy
| | - Mario Pirisi
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
- Immuno-Rheumatology Unit, Azienda Ospedaliero-Universitaria Maggiore della Carità, Novara, Italy
| | - Cristoforo Comi
- Interdisciplinary Research Center of Autoimmune Diseases (IRCAD), University of Piemonte Orientale, Novara, Italy
- Department of Translational Medicine, Neurology Unit, University of Piemonte Orientale, Novara, Italy
| |
Collapse
|
21
|
Groblewska M, Muszyński P, Wojtulewska-Supron A, Kulczyńska-Przybik A, Mroczko B. The Role of Visinin-Like Protein-1 in the Pathophysiology of Alzheimer's Disease. J Alzheimers Dis 2016; 47:17-32. [PMID: 26402751 DOI: 10.3233/jad-150060] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
Abstract
Calcium ions are crucial in the process of information transmission and integration in the central nervous system (CNS). These ions participate not only in intracellular mechanisms but also in intercellular processes. The changes in the concentration of Ca2 + ions modulate synaptic transmission, whereas neuronal activity induces calcium ion waves. Disturbed calcium homeostasis is thought to be one of the main features in the pathophysiology of Alzheimer's disease (AD), and AD pathogenesis is closely connected to Ca2 + signaling pathways. The effects of changes in neuronal Ca2 + are mediated by neuronal calcium sensor (NCS) proteins. It has been revealed that NCS proteins, with special attention to visinin-like protein 1 (VILIP-1), might have a connection to the etiology of AD. In the CNS, VILIP-1 influences the intracellular neuronal signaling pathways involved in synaptic plasticity, such as cyclic nucleotide cascades and nicotinergic signaling. This particular protein is implicated in calcium-mediated neuronal injury as well. VILIP-1 also participates in the pathological mechanisms of altered Ca2 + homeostasis, leading to neuronal loss. These findings confirm the utility of VILIP-1 as a useful biomarker of neuronal injury. Moreover, VILIP-1 plays a vital role in linking calcium-mediated neurotoxicity and AD-type pathological changes. The disruption of Ca2 + homeostasis caused by AD-type neurodegeneration may result in the damage of VILIP-1-containing neurons in the brain, leading to increased cerebrospinal fluid levels of VILIP-1. Thus, the aim of this overview is to describe the relationships of the NCS protein VILIP-1 with the pathogenetic factors of AD and neurodegenerative processes, as well as its potential clinical usefulness as a biomarker of AD. Moreover, we describe the current and probable therapeutic strategies for AD, targeting calcium-signaling pathways and VILIP-1.
Collapse
Affiliation(s)
| | - Paweł Muszyński
- Department of Neurodegeneration Diagnostics, Medical University of Białystok, Poland
| | | | | | - Barbara Mroczko
- Department of Biochemical Diagnostics, University Hospital in Białystok, Poland.,Department of Neurodegeneration Diagnostics, Medical University of Białystok, Poland
| |
Collapse
|
22
|
Penazzi L, Bakota L, Brandt R. Microtubule Dynamics in Neuronal Development, Plasticity, and Neurodegeneration. INTERNATIONAL REVIEW OF CELL AND MOLECULAR BIOLOGY 2015; 321:89-169. [PMID: 26811287 DOI: 10.1016/bs.ircmb.2015.09.004] [Citation(s) in RCA: 67] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Neurons are the basic information-processing units of the nervous system. In fulfilling their task, they establish a structural polarity with an axon that can be over a meter long and dendrites with a complex arbor, which can harbor ten-thousands of spines. Microtubules and their associated proteins play important roles during the development of neuronal morphology, the plasticity of neurons, and neurodegenerative processes. They are dynamic structures, which can quickly adapt to changes in the environment and establish a structural scaffold with high local variations in composition and stability. This review presents a comprehensive overview about the role of microtubules and their dynamic behavior during the formation and maturation of processes and spines in the healthy brain, during aging and under neurodegenerative conditions. The review ends with a discussion of microtubule-targeted therapies as a perspective for the supportive treatment of neurodegenerative disorders.
Collapse
Affiliation(s)
- Lorène Penazzi
- Department of Neurobiology, University of Osnabrück, Osnabrück, Germany
| | - Lidia Bakota
- Department of Neurobiology, University of Osnabrück, Osnabrück, Germany
| | - Roland Brandt
- Department of Neurobiology, University of Osnabrück, Osnabrück, Germany
| |
Collapse
|
23
|
Rojanathammanee L, Floden AM, Manocha GD, Combs CK. Attenuation of microglial activation in a mouse model of Alzheimer's disease via NFAT inhibition. J Neuroinflammation 2015; 12:42. [PMID: 25889879 PMCID: PMC4355356 DOI: 10.1186/s12974-015-0255-2] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 01/25/2015] [Indexed: 12/21/2022] Open
Abstract
Background Amyloid β (Aβ) peptide is hypothesized to stimulate microglia to acquire their characteristic proinflammatory phenotype in Alzheimer’s disease (AD) brains. The specific mechanisms by which Aβ leads to microglial activation remain an area of interest for identifying attractive molecular targets for intervention. Based upon the fact that microglia express the proinflammatory transcription factor, nuclear factor of activated T cells (NFAT), we hypothesized that NFAT activity is required for the Aβ-stimulated microgliosis that occurs during disease. Methods Primary murine microglia cultures were stimulated with Aβ in the absence or presence of NFAT inhibitors, FK506 and tat-VIVIT peptide, to quantify secretion of cytokines, neurotoxins, or Aβ phagocytosis. A transgenic mouse model of AD, APP/PS1, was treated subcutaneously via mini-osmotic pumps with FK506 or tat-VIVIT to quantify effects on cytokines, microgliosis, plaque load, and memory. Results Expression of various NFAT isoforms was verified in primary murine microglia through Western blot analysis. Microglial cultures were stimulated with Aβ fibrils in the absence or presence of the NFAT inhibitors, FK506 and tat-VIVIT, to demonstrate that NFAT activity regulated Aβ phagocytosis, neurotoxin secretion, and cytokine secretion. Delivery of FK506 and tat-VIVIT to transgenic APP/PS1 mice attenuated spleen but not brain cytokine levels. However, FK506 and tat-VIVIT significantly attenuated both microgliosis and Aβ plaque load in treated mice compared to controls. Surprisingly, this did not correlate with changes in memory performance via T-maze testing. Conclusions Our findings suggest that development of specific NFAT inhibitors may offer promise as an effective strategy for attenuating the microgliosis and Aβ plaque deposition that occur in AD. Electronic supplementary material The online version of this article (doi:10.1186/s12974-015-0255-2) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Lalida Rojanathammanee
- Institute of Science, Suranaree University of Technology, 111 University Avenue, Suranaree Subdistric, Nakhon Ratchasima, 30000, Thailand. .,Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, 504 Hamline Street, Neuroscience Building, Grand Forks, ND, 58203, USA.
| | - Angela M Floden
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, 504 Hamline Street, Neuroscience Building, Grand Forks, ND, 58203, USA.
| | - Gunjan D Manocha
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, 504 Hamline Street, Neuroscience Building, Grand Forks, ND, 58203, USA.
| | - Colin K Combs
- Department of Basic Sciences, University of North Dakota School of Medicine and Health Sciences, 504 Hamline Street, Neuroscience Building, Grand Forks, ND, 58203, USA.
| |
Collapse
|
24
|
Nimodipine enhances neurite outgrowth in dopaminergic brain slice co-cultures. Int J Dev Neurosci 2014; 40:1-11. [PMID: 25447789 DOI: 10.1016/j.ijdevneu.2014.10.005] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/19/2014] [Revised: 10/24/2014] [Accepted: 10/26/2014] [Indexed: 11/24/2022] Open
Abstract
Calcium ions (Ca(2+)) play important roles in neuroplasticity and the regeneration of nerves. Intracellular Ca(2+) concentrations are regulated by Ca(2+) channels, among them L-type voltage-gated Ca(2+) channels, which are inhibited by dihydropyridines like nimodipine. The purpose of this study was to investigate the effect of nimodipine on neurite growth during development and regeneration. As an appropriate model to study neurite growth, we chose organotypic brain slice co-cultures of the mesocortical dopaminergic projection system, consisting of the ventral tegmental area/substantia nigra and the prefrontal cortex from neonatal rat brains. Quantification of the density of the newly built neurites in the border region (region between the two cultivated slices) of the co-cultures revealed a growth promoting effect of nimodipine at concentrations of 0.1μM and 1μM that was even more pronounced than the effect of the growth factor NGF. This beneficial effect was absent when 10μM nimodipine were applied. Toxicological tests revealed that the application of nimodipine at this higher concentration slightly induced caspase 3 activation in the cortical part of the co-cultures, but did neither affect the amount of lactate dehydrogenase release or propidium iodide uptake nor the ratio of bax/bcl-2. Furthermore, the expression levels of different genes were quantified after nimodipine treatment. The expression of Ca(2+) binding proteins, immediate early genes, glial fibrillary acidic protein, and myelin components did not change significantly after treatment, indicating that the regulation of their expression is not primarily involved in the observed nimodipine mediated neurite growth. In summary, this study revealed for the first time a neurite growth promoting effect of nimodipine in the mesocortical dopaminergic projection system that is highly dependent on the applied concentrations.
Collapse
|
25
|
Swart C, Haylett W, Kinnear C, Johnson G, Bardien S, Loos B. Neurodegenerative disorders: dysregulation of a carefully maintained balance? Exp Gerontol 2014; 58:279-91. [PMID: 25219768 DOI: 10.1016/j.exger.2014.09.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/17/2014] [Revised: 09/01/2014] [Accepted: 09/08/2014] [Indexed: 10/24/2022]
Abstract
The aggregation of misfolded proteins has long been regarded as a pathological event in neurodegenerative diseases such as Alzheimer's disease, Parkinson's disease and Huntington's disease. However, the exact molecular mechanisms that govern protein metabolism that may lead to toxicity remain largely unclear. Originally targeted as the causative agent, it has since become evident that aggregation formation may not be necessary for disease progression and studies show that they may even serve functional and protective roles. Although the focus has since shifted to the toxicity of intermediate protein species preceding aggregation formation, many questions remain: Is the blame for the neural destruction to be put on one event alone, or rather on a state of cellular disequilibrium resulting from multiple events? If the cause is multifactorial, then what triggers the toxic cascade and how can this be targeted therapeutically? In order to understand the origin of toxicity, the exact underlying mechanism and impact of each contributing process must be assessed. Therefore, the structural properties, mechanism of formation, cytotoxic and/or protective effects, as well as the clinical impact of protein intermediates and aggregates will be reviewed here with the goal to establish a neurodegenerative disease model aimed at improving current therapeutics, which may ultimately contribute towards improved treatment modalities.
Collapse
Affiliation(s)
- Chrisna Swart
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa.
| | - William Haylett
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Craig Kinnear
- South African Medical Research Council Centre for Tuberculosis Research, Cape Town, South Africa
| | - Glynis Johnson
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Soraya Bardien
- Division of Molecular Biology and Human Genetics, Faculty of Medicine and Health Sciences, Stellenbosch University, Cape Town, South Africa
| | - Ben Loos
- Department of Physiological Sciences, Faculty of Science, Stellenbosch University, Cape Town, South Africa.
| |
Collapse
|
26
|
Nimmrich V, Eckert A. Calcium channel blockers and dementia. Br J Pharmacol 2014; 169:1203-10. [PMID: 23638877 DOI: 10.1111/bph.12240] [Citation(s) in RCA: 111] [Impact Index Per Article: 10.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2012] [Revised: 04/15/2013] [Accepted: 04/22/2013] [Indexed: 12/25/2022] Open
Abstract
Degenerative dementia is mainly caused by Alzheimer's disease and/or cerebrovascular abnormalities. Disturbance of the intracellular calcium homeostasis is central to the pathophysiology of neurodegeneration. In Alzheimer's disease, enhanced calcium load may be brought about by extracellular accumulation of amyloid-β. Recent studies suggest that soluble forms facilitate influx through calcium-conducting ion channels in the plasma membrane, leading to excitotoxic neurodegeneration. Calcium channel blockade attenuates amyloid-β-induced neuronal decline in vitro and is neuroprotective in animal models. Vascular dementia, on the other hand, is caused by cerebral hypoperfusion and may benefit from calcium channel blockade due to relaxation of the cerebral vasculature. Several calcium channel blockers have been tested in clinical trials of dementia and the outcome is heterogeneous. Nimodipine as well as nilvadipine prevent cognitive decline in some trials, whereas other calcium channel blockers failed. In trials with a positive outcome, BP reduction did not seem to play a role in preventing dementia, indicating a direct protecting effect on neurons. An optimization of calcium channel blockers for the treatment of dementia may involve an increase of selectivity for presynaptic calcium channels and an improvement of the affinity to the inactivated state. Novel low molecular weight compounds suitable for proof-of-concept studies are now available.
Collapse
Affiliation(s)
- V Nimmrich
- Neuroscience Research, GPRD, AbbVie GmbH, Ludwigshafen, Germany.
| | | |
Collapse
|
27
|
Does Na⁺/Ca²⁺ exchanger, NCX, represent a new druggable target in stroke intervention? Transl Stroke Res 2013; 5:145-55. [PMID: 24323727 DOI: 10.1007/s12975-013-0308-8] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2013] [Revised: 10/15/2013] [Accepted: 11/06/2013] [Indexed: 12/22/2022]
Abstract
Stroke causes a rapid cell death in the core of the injured region and triggers mechanisms in surrounding penumbra area that leads to changes in concentrations of several ions like intracellular Ca²⁺, Na⁺, H⁺, K⁺, and radicals such as reactive oxygen species and reactive nitrogen species. When a dysregulation of homeostasis of these messengers occurs, it can trigger cell death. In particular, it is widely accepted that a critical factor in determining neuronal death during cerebral ischemia is progressive dysregulation of Ca²⁺, Na⁺, K⁺, and H⁺ homeostasis that activate several death pathways, including oxidative and nitrosative stress, mitochondrial dysfunction, protease activation, and apoptosis. In the last decade, several seminal experimental works are markedly changing the scenario of research of principal players of an ischemic event. Indeed, some plasma membrane channels and transporters, involved in the control of Ca²⁺, Na⁺, K⁺, and H⁺ ion influx or efflux and, therefore, responsible for maintaining the homeostasis of these four cations, might function as crucial players in initiation of brain ischemic process. Indeed, these proteins, by regulating ionic homeostasis, may provide the molecular basis underlying glutamate-independent Ca²⁺ and Na⁺ overload mechanisms in neuronal ischemic cell death and, most importantly, may represent more suitable molecular targets for therapeutic intervention. Recently, a great deal of interest has been devoted to clarify the role of the plasma membrane protein known as Na⁺/Ca²⁺ exchanger, a transporter able to control Na⁺ and Ca²⁺ homeostasis. In this review, the pathophysiological role of NCX and its implication as a potential target in stroke intervention will be examined.
Collapse
|
28
|
Cataldi M. The changing landscape of voltage-gated calcium channels in neurovascular disorders and in neurodegenerative diseases. Curr Neuropharmacol 2013; 11:276-97. [PMID: 24179464 PMCID: PMC3648780 DOI: 10.2174/1570159x11311030004] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2012] [Revised: 02/02/2013] [Accepted: 02/14/2013] [Indexed: 12/12/2022] Open
Abstract
It is a common belief that voltage-gated calcium channels (VGCC) cannot carry toxic amounts of Ca2+ in neurons. Also, some of them as L-type channels are essential for Ca2+-dependent regulation of prosurvival gene-programs. However, a wealth of data show a beneficial effect of drugs acting on VGCCs in several neurodegenerative and neurovascular diseases. In the present review, we explore several mechanisms by which the “harmless” VGCCs may become “toxic” for neurons. These mechanisms could explain how, though usually required for neuronal survival, VGCCs may take part in neurodegeneration. We will present evidence showing that VGCCs can carry toxic Ca2+ when: a) their density or activity increases because of aging, chronic hypoxia or exposure to β-amyloid peptides or b) Ca2+-dependent action potentials carry high Ca2+ loads in pacemaker neurons. Besides, we will examine conditions in which VGCCs promote neuronal cell death without carrying excess Ca2+. This can happen, for instance, when they carry metal ions into the neuronal cytoplasm or when a pathological decrease in their activity weakens Ca2+-dependent prosurvival gene programs. Finally, we will explore the role of VGCCs in the control of nonneuronal cells that take part to neurodegeneration like those of the neurovascular unit or of microglia.
Collapse
Affiliation(s)
- Mauro Cataldi
- Division of Pharmacology, Department of Neuroscience, Reproductive and Odontostomatological Sciences, Federico II University of Naples, Italy
| |
Collapse
|
29
|
Sanz JM, Chiozzi P, Colaianna M, Zotti M, Ferrari D, Trabace L, Zuliani G, Di Virgilio F. Nimodipine inhibits IL-1β release stimulated by amyloid β from microglia. Br J Pharmacol 2012; 167:1702-11. [PMID: 22831460 PMCID: PMC3525872 DOI: 10.1111/j.1476-5381.2012.02112.x] [Citation(s) in RCA: 44] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2012] [Revised: 07/09/2012] [Indexed: 10/28/2022] Open
Abstract
BACKGROUND AND PURPOSE There is growing evidence that inflammation plays a major role in the pathogenesis of neural damage caused by deposition of amyloid β (Aβ) in the brain. Nimodipine has received attention as a drug that might improve learning and reduce cognitive deficits in Alzheimer's disease, but the mechanism of action is poorly known. In this study, we tested the hypothesis that nimodipine inhibited Aβ-stimulated IL-1β release from microglia. EXPERIMENTAL APPROACH Cultures of N13 microglia cells or primary mouse microglia were treated with nimodipine, and intracellular accumulation and release of IL-1β in response to Aβ or to the P2 receptor agonists ATP and benzoyl ATP (BzATP) were measured. Accumulation of IL-1β was measured in vivo after intrahippocampal inoculation of Aβ in the absence or presence of nimodipine. The effect of nimodipine on Aβ-triggered cytotoxicity was also investigated. KEY RESULTS We show here that nimodipine dose-dependently inhibited Aβ-stimulated IL-1β synthesis and release from primary microglia and microglia cell lines. Furthermore, nimodipine also inhibited Aβ-induced IL-1βin vivo accumulation at concentrations known to be reached in the CNS. Finally, nimodipine protected microglia from Aβ-dependent cytotoxicity. CONCLUSION AND IMPLICATIONS These data suggest that alleviation of symptoms of Alzheimer's disease following nimodipine administration might be due to an anti-inflammatory effect and point to a novel role for nimodipine as a centrally acting anti-inflammatory drug.
Collapse
Affiliation(s)
- JM Sanz
- Department of Clinical and Experimental Medicine, University of FerraraFerrara, Italy
| | - P Chiozzi
- Department of Experimental and Diagnostic Medicine, University of FerraraFerrara, Italy
| | - M Colaianna
- Department of Biomedical Sciences, University of FoggiaFoggia, Italy
| | - M Zotti
- Department of Biomedical Sciences, University of FoggiaFoggia, Italy
| | - D Ferrari
- Department of Experimental and Diagnostic Medicine, University of FerraraFerrara, Italy
| | - L Trabace
- Department of Biomedical Sciences, University of FoggiaFoggia, Italy
| | - G Zuliani
- Department of Clinical and Experimental Medicine, University of FerraraFerrara, Italy
| | - F Di Virgilio
- Department of Experimental and Diagnostic Medicine, University of FerraraFerrara, Italy
| |
Collapse
|
30
|
Inhibition of human astrocyte and microglia neurotoxicity by calcium channel blockers. Neuropharmacology 2012; 63:685-91. [PMID: 22659089 DOI: 10.1016/j.neuropharm.2012.05.033] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/14/2012] [Revised: 05/12/2012] [Accepted: 05/24/2012] [Indexed: 11/24/2022]
Abstract
We examined the effects of L-type calcium channel blockers (CCBs) on toxicity exerted by activated human astrocytes and microglia towards SH-SY5Y human neuronal cells. The CCBs nimodipine (NDP) and verapamil (VPM) both significantly suppressed toxic secretions from human astrocytes and astrocytoma U-373 MG cells that were induced by interferon (IFN)-γ. NDP also inhibited neurotoxic secretions of human microglia and monocytic THP-1 cells that were induced by the combination of lipopolysaccharide and IFN-γ. In human astrocytes, both NDP and VPM reduced IFN-γ-induced phosphorylation of signal transducer and activator of transcription (STAT) 3. They also inhibited the astrocytic production of IFN-γ-inducible T cell α chemoattractant (I-TAC). These results suggest that CCBs attenuate IFN-γ-induced neurotoxicity of human astrocytes through inhibition of the STAT3 signaling pathway. L-type CCBs, especially NDP, might be a useful treatment option for a broad spectrum of neurodegenerative diseases, including Alzheimer disease, where the pathology is believed to be exacerbated by neurotoxic glial activation.
Collapse
|
31
|
Kagan BL, Jang H, Capone R, Arce FT, Ramachandran S, Lal R, Nussinov R. Antimicrobial properties of amyloid peptides. Mol Pharm 2012; 9:708-17. [PMID: 22081976 PMCID: PMC3297685 DOI: 10.1021/mp200419b] [Citation(s) in RCA: 174] [Impact Index Per Article: 13.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
More than two dozen clinical syndromes known as amyloid diseases are characterized by the buildup of extended insoluble fibrillar deposits in tissues. These amorphous Congo red staining deposits known as amyloids exhibit a characteristic green birefringence and cross-β structure. Substantial evidence implicates oligomeric intermediates of amyloids as toxic species in the pathogenesis of these chronic disease states. A growing body of data has suggested that these toxic species form ion channels in cellular membranes causing disruption of calcium homeostasis, membrane depolarization, energy drainage, and in some cases apoptosis. Amyloid peptide channels exhibit a number of common biological properties including the universal U-shape β-strand-turn-β-strand structure, irreversible and spontaneous insertion into membranes, production of large heterogeneous single-channel conductances, relatively poor ion selectivity, inhibition by Congo red, and channel blockade by zinc. Recent evidence has suggested that increased amounts of amyloids not only are toxic to its host target cells but also possess antimicrobial activity. Furthermore, at least one human antimicrobial peptide, protegrin-1, which kills microbes by a channel-forming mechanism, has been shown to possess the ability to form extended amyloid fibrils very similar to those of classic disease-forming amyloids. In this paper, we will review the reported antimicrobial properties of amyloids and the implications of these discoveries for our understanding of amyloid structure and function.
Collapse
Affiliation(s)
- Bruce L. Kagan
- Department of Psychiatry, David Geffen School of Medicine, Semel Institute for Neuroscience & Human Behavior, University of California, Los Angeles, California 90024, U.S.A
| | - Hyunbum Jang
- Center for Cancer Research Nanobiology Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland 21702, U.S.A
| | - Ricardo Capone
- Departments of Bioengineering and of Mechanical and Aerospace Engineering and Material Science Program, University of California, San Diego, La Jolla, California 92093, U.S.A
| | - Fernando Teran Arce
- Departments of Bioengineering and of Mechanical and Aerospace Engineering and Material Science Program, University of California, San Diego, La Jolla, California 92093, U.S.A
| | - Srinivasan Ramachandran
- Departments of Bioengineering and of Mechanical and Aerospace Engineering and Material Science Program, University of California, San Diego, La Jolla, California 92093, U.S.A
| | - Ratnesh Lal
- Departments of Bioengineering and of Mechanical and Aerospace Engineering and Material Science Program, University of California, San Diego, La Jolla, California 92093, U.S.A
| | - Ruth Nussinov
- Center for Cancer Research Nanobiology Program, SAIC-Frederick, Inc., NCI-Frederick, Frederick, Maryland 21702, U.S.A
- Department of Human Molecular Genetics and Biochemistry, Sackler School of Medicine, Tel Aviv University, Tel Aviv 69978, Israel
| |
Collapse
|
32
|
Thibault O, Pancani T, Landfield PW, Norris CM. Reduction in neuronal L-type calcium channel activity in a double knock-in mouse model of Alzheimer's disease. BIOCHIMICA ET BIOPHYSICA ACTA 2012; 1822:546-9. [PMID: 22265986 PMCID: PMC3293940 DOI: 10.1016/j.bbadis.2012.01.004] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/31/2011] [Revised: 12/20/2011] [Accepted: 01/04/2012] [Indexed: 11/25/2022]
Abstract
Increased function of neuronal L-type voltage-sensitive Ca(2+) channels (L-VSCCs) is strongly linked to impaired memory and altered hippocampal synaptic plasticity in aged rats. However, no studies have directly assessed L-VSCC function in any of the common mouse models of Alzheimer's disease where neurologic deficits are typically more robust. Here, we used cell-attached patch-clamp recording techniques to measure L-VSCC activity in CA1 pyramidal neurons of partially dissociated hippocampal "zipper" slices prepared from 14-month-old wild-type mice and memory-impaired APP/PS1 double knock-in mice. Surprisingly, the functional channel density of L-VSCCs was significantly reduced in the APP/PS1 group. No differences in voltage dependency and unitary conductance of L-VSCCs were observed. The results suggest that mechanisms for Ca(2+) dysregulation can differ substantially between animal models of normal aging and models of pathological aging.
Collapse
Affiliation(s)
- Olivier Thibault
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, KY 40536 USA
| | - Tristano Pancani
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, KY 40536 USA
| | - Philip W. Landfield
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, KY 40536 USA
| | - Christopher M. Norris
- Department of Molecular and Biomedical Pharmacology, University of Kentucky College of Medicine, Lexington, KY 40536 USA
- Sanders-Brown Center on Aging, University of Kentucky College of Medicine, Lexington, KY 40536 USA
| |
Collapse
|
33
|
Uteshev VV. α7 nicotinic ACh receptors as a ligand-gated source of Ca(2+) ions: the search for a Ca(2+) optimum. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 740:603-38. [PMID: 22453962 DOI: 10.1007/978-94-007-2888-2_27] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
The spatiotemporal distribution of cytosolic Ca(2+) ions is a key determinant of neuronal behavior and survival. Distinct sources of Ca(2+) ions including ligand- and voltage-gated Ca(2+) channels contribute to intracellular Ca(2+) homeostasis. Many normal physiological and therapeutic neuronal functions are Ca(2+)-dependent, however an excess of cytosolic Ca(2+) or a lack of the appropriate balance between Ca(2+) entry and clearance may destroy cellular integrity and cause cellular death. Therefore, the existence of optimal spatiotemporal patterns of cytosolic Ca(2+) elevations and thus, optimal activation of ligand- and voltage-gated Ca(2+) ion channels are postulated to benefit neuronal function and survival. Alpha7 nicotinic -acetylcholine receptors (nAChRs) are highly permeable to Ca(2+) ions and play an important role in modulation of neurotransmitter release, gene expression and neuroprotection in a variety of neuronal and non-neuronal cells. In this review, the focus is placed on α7 nAChR-mediated currents and Ca(2+) influx and how this source of Ca(2+) entry compares to NMDA receptors in supporting cytosolic Ca(2+) homeostasis, neuronal function and survival.
Collapse
Affiliation(s)
- Victor V Uteshev
- Department of Pharmacology & Neuroscience, University of North Texas Health Science Center, 3500 Camp Bowie Blvd, Fort Worth, TX 76107, USA.
| |
Collapse
|
34
|
Kagan BL. Membrane pores in the pathogenesis of neurodegenerative disease. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2012; 107:295-325. [PMID: 22482454 DOI: 10.1016/b978-0-12-385883-2.00001-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
The neurodegenerative diseases described in this volume, as well as many nonneurodegenerative diseases, are characterized by deposits known as amyloid. Amyloid has long been associated with these various diseases as a pathological marker and has been implicated directly in the molecular pathogenesis of disease. However, increasing evidence suggests that these proteinaceous Congo red staining deposits may not be toxic or destructive of tissue. Recent studies strongly implicate smaller aggregates of amyloid proteins as the toxic species underlying these neurodegenerative diseases. Despite the outward obvious differences among these clinical syndromes, there are some striking similarities in their molecular pathologies. These include dysregulation of intracellular calcium levels, impairment of mitochondrial function, and the ability of virtually all amyloid peptides to form ion-permeable pores in lipid membranes. Pore formation is enhanced by environmental factors that promote protein aggregation and is inhibited by agents, such as Congo red, which prevent aggregation. Remarkably, the pores formed by a variety of amyloid peptides from neurodegenerative and other diseases share a common set of physiologic properties. These include irreversible insertion of the pores in lipid membranes, formation of heterodisperse pore sizes, inhibition by Congo red of pore formation, blockade of pores by zinc, and a relative lack of ion selectivity and voltage dependence. Although there exists some information about the physical structure of these pores, molecular modeling suggests that 4-6-mer amyloid subunits may assemble into 24-mer pore-forming aggregates. The molecular structure of these pores may resemble the β-barrel structure of the toxics pore formed by bacterial toxins, such as staphylococcal α-hemolysin, anthrax toxin, and Clostridium perfringolysin.
Collapse
Affiliation(s)
- Bruce L Kagan
- Department of Psychiatry & Biobehavioral Sciences, David Geffen School of Medicine at UCLA, Semel Institute for Neuroscience and Human Behavior, Los Angeles, California, USA
| |
Collapse
|
35
|
Copenhaver PF, Anekonda TS, Musashe D, Robinson KM, Ramaker JM, Swanson TL, Wadsworth TL, Kretzschmar D, Woltjer RL, Quinn JF. A translational continuum of model systems for evaluating treatment strategies in Alzheimer's disease: isradipine as a candidate drug. Dis Model Mech 2011; 4:634-48. [PMID: 21596710 PMCID: PMC3180227 DOI: 10.1242/dmm.006841] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2010] [Accepted: 04/18/2011] [Indexed: 01/16/2023] Open
Abstract
A growing body of evidence supports the 'calcium hypothesis' of Alzheimer's disease (AD), which postulates that a variety of insults might disrupt the homeostatic regulation of neuronal calcium (Ca(2+)) in the brain, resulting in the progressive symptoms that typify the disease. However, despite ongoing efforts to develop new methods for testing therapeutic compounds that might be beneficial in AD, no single bioassay permits both rapid screening and in vivo validation of candidate drugs that target specific components of the Ca(2+) regulatory machinery. To address this issue, we have integrated four distinct model systems that provide complementary information about a trial compound: the human neuroblastoma MC65 line, which provides an in vitro model of amyloid toxicity; a transgenic Drosophila model, which develops age-dependent pathologies associated with AD; the 3×TgAD transgenic mouse, which recapitulates many of the neuropathological features that typify AD; and the embryonic nervous system of Manduca, which provides a novel in vivo assay for the acute effects of amyloid peptides on neuronal motility. To demonstrate the value of this 'translational suite' of bioassays, we focused on a set of clinically approved dihydropyridines (DHPs), a class of well-defined inhibitors of L-type calcium channels that have been suggested to be neuroprotective in AD. Among the DHPs tested in this study, we found that isradipine reduced the neurotoxic consequences of β-amyloid accumulation in all four model systems without inducing deleterious side effects. Our results provide new evidence in support of the Ca(2+) hypothesis of AD, and indicate that isradipine represents a promising drug for translation into clinical trials. In addition, these studies also demonstrate that this continuum of bioassays (representing different levels of complexity) provides an effective means of evaluating other candidate compounds that target specific components of the Ca(2+) regulatory machinery and that therefore might be beneficial in the treatment of AD.
Collapse
Affiliation(s)
- Philip F Copenhaver
- Department of Cell and Developmental Biology, Oregon Health andScience University, 3181 SW Sam Jackson Park Road, Portland, OR 97239, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Kawahara M, Ohtsuka I, Yokoyama S, Kato-Negishi M, Sadakane Y. Membrane Incorporation, Channel Formation, and Disruption of Calcium Homeostasis by Alzheimer's β-Amyloid Protein. Int J Alzheimers Dis 2011; 2011:304583. [PMID: 21547225 PMCID: PMC3087492 DOI: 10.4061/2011/304583] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/16/2010] [Revised: 12/22/2010] [Accepted: 01/27/2011] [Indexed: 01/09/2023] Open
Abstract
Oligomerization, conformational changes, and the consequent neurodegeneration of Alzheimer's β-amyloid protein (AβP) play crucial roles in the pathogenesis of Alzheimer's disease (AD). Mounting evidence suggests that oligomeric AβPs cause the disruption of calcium homeostasis, eventually leading to neuronal death. We have demonstrated that oligomeric AβPs directly incorporate into neuronal membranes, form cation-sensitive ion channels (“amyloid channels”), and cause the disruption of calcium homeostasis via the amyloid channels. Other disease-related amyloidogenic proteins, such as prion protein in prion diseases or α-synuclein in dementia with Lewy bodies, exhibit similarities in the incorporation into membranes and the formation of calcium-permeable channels. Here, based on our experimental results and those of numerous other studies, we review the current understanding of the direct binding of AβP into membrane surfaces and the formation of calcium-permeable channels. The implication of composition of membrane lipids and the possible development of new drugs by influencing membrane properties and attenuating amyloid channels for the treatment and prevention of AD is also discussed.
Collapse
Affiliation(s)
- Masahiro Kawahara
- Department of Analytical Chemistry, School of Pharmaceutical Sciences, Kyushu University of Health and Welfare, 1714-1 Yoshino-cho, Nobeoka-shi, Miyazaki 882-8508, Japan
| | | | | | | | | |
Collapse
|
37
|
Herrera JL, Diaz M, Hernández-Fernaud JR, Salido E, Alonso R, Fernández C, Morales A, Marin R. Voltage-dependent anion channel as a resident protein of lipid rafts: post-transductional regulation by estrogens and involvement in neuronal preservation against Alzheimer's disease. J Neurochem 2011; 116:820-7. [PMID: 21214547 DOI: 10.1111/j.1471-4159.2010.06987.x] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
The voltage-dependent anion channel, VDAC, is present at the neuronal membrane, where it appears to participate, among others, in the extrinsic apoptotic pathway and in the modulation of amyloid-beta induced injury, suggesting the involvement of this channel in Alzheimer's disease (AD) neurotoxicity. VDAC is also highly concentrated in neuronal lipid raft microdomains of different mouse and human cognitive areas, where it has been shown associated with estrogen receptor alpha (ERα), as a part of a `signalosome' that may activate some intracellular signal transduction. At the plasma membrane level, estrogens and antiestrogens (tamoxifen) have been demonstrated to exert rapid antagonist effects on the activation of VDAC, through their distinct effects on the channel post-transductional modulation. Therefore, part of the alternative mechanisms of estrogen related to neuroprotection against amyloid-beta may involve VDAC phosphorylation, in order to maintain the channel in an unactivated (closing) state. Interestingly, VDAC-ERα association has been shown to be disrupted in neuronal lipid rafts of AD brains, in correlation with the aberrant lipid composition observed in these microstructures, suggesting that disturbance of protein interactions may be related to variation in the physico-chemical properties of these microdomains.
Collapse
Affiliation(s)
- Jose Luis Herrera
- Department of Physiology, School of Medicine, La Laguna University, Tenerife, Spain
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Kosenko E, Poghosyan A, Kaminsky Y. Subcellular compartmentalization of proteolytic enzymes in brain regions and the effects of chronic β-amyloid treatment. Brain Res 2011; 1369:184-93. [DOI: 10.1016/j.brainres.2010.10.078] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2010] [Revised: 10/19/2010] [Accepted: 10/20/2010] [Indexed: 12/14/2022]
|
39
|
Lopez JR, Shtifman A. Intracellular β-amyloid accumulation leads to age-dependent progression of Ca2+ dysregulation in skeletal muscle. Muscle Nerve 2010; 42:731-8. [PMID: 20665519 DOI: 10.1002/mus.21745] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
Intramyofiber accumulation of β-amyloid fragments (Aβ) is a pathologic hallmark of inclusion-body myositis (IBM), a progressive skeletal muscle disorder. We investigated the temporal pattern of alterations in the resting cytoplasmic [Ca(2+)] ([Ca(2+)](i)) as well as the depolarization-evoked Ca(2+) release from the sarcoplasmic reticulum in skeletal muscle from transgenic mice expressing human βAPP (MCK-βAPP). MCK-βAPP mice show an age-dependent increase in [Ca(2+)](i) along with a reduction in depolarization-evoked Ca(2+) release, which appear well before the other reported aspects of IBM, such as inclusion formation, inflammation, centralized nuclei, atrophy, and skeletal muscle weakness. In the young MCK-βAPP animals the increase in resting [Ca(2+)](i) can be attributed largely to Ca(2+) influx through nifedipine-sensitive Ca(2+) channels. In the adult MCK-βAPP mice, in addition to the nifedipine-sensitive pathway, there is also a substantial contribution by the intracellular compartments to the increase in [Ca(2+)](i). These results suggest that β-amyloid-induced disuption of Ca(2+) handling may represent an early event in the pathogenesis of IBM.
Collapse
Affiliation(s)
- Josè R Lopez
- Department of Anesthesia, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | |
Collapse
|
40
|
Li L, Tsai HJ, Li L, Wang XM. Icariin inhibits the increased inward calcium currents induced by amyloid-beta(25-35) peptide in CA1 pyramidal neurons of neonatal rat hippocampal slice. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2010; 38:113-25. [PMID: 20128049 DOI: 10.1142/s0192415x10007701] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Overload of intracellular calcium caused by amyloid-beta peptide has been implicated in the pathogenesis of neuronal damage in Alzheimer's disease. Voltage-gated calcium channels (VGCCs) provide one of the major sources of Ca(2+) entry into cells. Here, we investigated whether icariin had effect on the changes of calcium currents induced by Abeta(25-35) in hippocampal pyramidal neurons. Using whole-cell patch-clamp, we showed that Abeta(25-35) enhanced the inward Ba(2+) and Ca(2+) currents. The currents were partially inhibited by Ni(2+) and completely suppressed by Cd(2+), indicating that Abeta(25-35) disrupts intracellular calcium homeostasis via the modulation of both L- and T-type channels. Furthermore, icariin nearly complete suppressed the abnormal inward calcium currents induced by Abeta(25-35) in a dose-dependant manner. Our findings suggest that the potential neuroprotective effect of icariin on Abeta(25-35)-induced neurotoxicity via the balance intracelluar calcium homeostasis.
Collapse
Affiliation(s)
- Li Li
- Department of Integrated Chinese and Western Medicine, First Hospital, Peking University, Beijing, China
| | | | | | | |
Collapse
|
41
|
Kaminsky YG, Marlatt MW, Smith MA, Kosenko EA. Subcellular and metabolic examination of amyloid-beta peptides in Alzheimer disease pathogenesis: evidence for Abeta(25-35). Exp Neurol 2009; 221:26-37. [PMID: 19751725 DOI: 10.1016/j.expneurol.2009.09.005] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2009] [Revised: 09/03/2009] [Accepted: 09/04/2009] [Indexed: 11/28/2022]
Abstract
Amyloid-beta peptide (Abeta) is a central player in the pathogenesis and diagnosis of Alzheimer disease. It aggregates to form the core of Alzheimer disease-associated plaques found in coordination with tau deposits in diseased individuals. Despite this clinical relevance, no single hypothesis satisfies and explicates the role of Abeta in toxicity and progression of the disease. To explore this area, investigators have focused on mechanisms of cellular dysfunction, aggregation, and maladaptive responses. Extensive research has been conducted using various methodologies to investigate Abeta peptides and oligomers, and these multiple facets have provided a wealth of data from specific models. Notably, the utility of each experiment must be considered in regards to the brain environment. The use of Abeta(25-35) in studies of cellular dysfunction has provided data indicating that the peptide is indeed responsible for multiple disturbances to cellular integrity. We will review how Abeta peptide induces oxidative stress and calcium homeostasis, and how multiple enzymes are deleteriously impacted by Abeta(25-35). Understanding and discussing the origin and properties of Abeta peptides is essential to evaluating their effects on various intracellular metabolic processes. Attention will also be specifically directed to metabolic compartmentation in affected brain cells, including mitochondrial, cytosolic, nuclear, and lysosomal enzymes.
Collapse
Affiliation(s)
- Yury G Kaminsky
- Institute of Theoretical and Experimental Biophysics, RAS, Pushchino, Russia.
| | | | | | | |
Collapse
|
42
|
Pike CJ, Carroll JC, Rosario ER, Barron AM. Protective actions of sex steroid hormones in Alzheimer's disease. Front Neuroendocrinol 2009; 30:239-58. [PMID: 19427328 PMCID: PMC2728624 DOI: 10.1016/j.yfrne.2009.04.015] [Citation(s) in RCA: 388] [Impact Index Per Article: 24.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/22/2009] [Revised: 04/25/2009] [Accepted: 04/28/2009] [Indexed: 12/19/2022]
Abstract
Risk for Alzheimer's disease (AD) is associated with age-related loss of sex steroid hormones in both women and men. In post-menopausal women, the precipitous depletion of estrogens and progestogens is hypothesized to increase susceptibility to AD pathogenesis, a concept largely supported by epidemiological evidence but refuted by some clinical findings. Experimental evidence suggests that estrogens have numerous neuroprotective actions relevant to prevention of AD, in particular promotion of neuron viability and reduction of beta-amyloid accumulation, a critical factor in the initiation and progression of AD. Recent findings suggest neural responsiveness to estrogen can diminish with age, reducing neuroprotective actions of estrogen and, consequently, potentially limiting the utility of hormone therapies in aged women. In addition, estrogen neuroprotective actions are also modulated by progestogens. Specifically, continuous progestogen exposure is associated with inhibition of estrogen actions whereas cyclic delivery of progestogens may enhance neural benefits of estrogen. In recent years, emerging literature has begun to elucidate a parallel relationship of sex steroid hormones and AD risk in men. Normal age-related testosterone loss in men is associated with increased risk to several diseases including AD. Like estrogen, testosterone has been established as an endogenous neuroprotective factor that not only increases neuronal resilience against AD-related insults, but also reduces beta-amyloid accumulation. Androgen neuroprotective effects are mediated both directly by activation of androgen pathways and indirectly by aromatization to estradiol and initiation of protective estrogen signaling mechanisms. The successful use of hormone therapies in aging men and women to delay, prevent, and or treat AD will require additional research to optimize key parameters of hormone therapy and may benefit from the continuing development of selective estrogen and androgen receptor modulators.
Collapse
Affiliation(s)
- Christian J Pike
- Davis School of Gerontology, University of Southern California, Los Angeles, CA 90089, USA.
| | | | | | | |
Collapse
|
43
|
Piacentini R, Ripoli C, Leone L, Misiti F, Clementi ME, D'Ascenzo M, Giardina B, Azzena GB, Grassi C. Role of methionine 35 in the intracellular Ca2+ homeostasis dysregulation and Ca2+-dependent apoptosis induced by amyloid beta-peptide in human neuroblastoma IMR32 cells. J Neurochem 2009; 107:1070-82. [PMID: 18990116 DOI: 10.1111/j.1471-4159.2008.05680.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/14/2023]
Abstract
Amyloid beta-peptide (Abeta) plays a fundamental role in the pathogenesis of Alzheimer's disease. We recently reported that the redox state of the methionine residue in position 35 of amyloid beta-peptide (Abeta) 1-42 (Met35) strongly affects the peptide's ability to trigger apoptosis and is thus a major determinant of its neurotoxicity. Dysregulation of intracellular Ca(2+) homeostasis resulting in the activation of pro-apoptotic pathways has been proposed as a mechanism underlying Abeta toxicity. Therefore, we investigated correlations between the Met35 redox state, Abeta toxicity, and altered intracellular Ca(2+) signaling in human neuroblastoma IMR32 cells. Cells incubated for 6-24 h with 10 microM Abeta1-42 exhibited significantly increased KCl-induced Ca(2+) transient amplitudes and resting free Ca(2+) concentrations. Nifedipine-sensitive Ca(2+) current densities and Ca(v)1 channel expression were markedly enhanced by Abeta1-42. None of these effects were observed when cells were exposed to Abeta containing oxidized Met35 (Abeta1-42(Met35-Ox)). Cell pre-treatment with the intracellular Ca(2+) chelator 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid acetoxymethyl ester (1 microM) or the Ca(v)1 channel blocker nifedipine (5 microM) significantly attenuated Abeta1-42-induced apoptosis but had no effect on Abeta1-42(Met35-Ox) toxicity. Collectively, these data suggest that reduced Met35 plays a critical role in Abeta1-42 toxicity by rendering the peptide capable of disrupting intracellular Ca(2+) homeostasis and thereby provoking apoptotic cell death.
Collapse
Affiliation(s)
- Roberto Piacentini
- Institute of Human Physiology, Medical School, Catholic University S Cuore, Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Shtifman A, Ward CW, Laver DR, Bannister ML, Lopez JR, Kitazawa M, LaFerla FM, Ikemoto N, Querfurth HW. Amyloid-β protein impairs Ca2+ release and contractility in skeletal muscle. Neurobiol Aging 2008; 31:2080-90. [PMID: 19108934 DOI: 10.1016/j.neurobiolaging.2008.11.003] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2008] [Revised: 11/03/2008] [Accepted: 11/07/2008] [Indexed: 11/18/2022]
Abstract
Inclusion body myositis (IBM), the most common muscle disorder in the elderly, is partly characterized by dysregulation of β-amyloid precursor protein (βAPP) expression and abnormal, intracellular accumulation of full-length βAPP and β-amyloid epitopes. The present study examined the effects of β-amyloid accumulation on force generation and Ca(2+) release in skeletal muscle from transgenic mice harboring human βAPP and assessed the consequence of Aβ(1-42) modulation of the ryanodine receptor Ca(2+) release channels (RyRs). β-Amyloid laden muscle produced less peak force and exhibited Ca(2+) transients with smaller amplitude. To determine whether modification of RyRs by β-amyloid underlie the effects observed in muscle, in vitro Ca(2+) release assays and RyR reconstituted in planar lipid bilayer experiments were conducted in the presence of Aβ(1-42). Application of Aβ(1-42) to RyRs in bilayers resulted in an increased channel open probability and changes in gating kinetics, while addition of Aβ(1-42) to the rabbit SR vesicles resulted in RyR-mediated Ca(2+) release. These data may relate altered βAPP metabolism in IBM to reductions in RyR-mediated Ca(2+) release and muscle contractility.
Collapse
Affiliation(s)
- Alexander Shtifman
- Department of Neurology, Caritas St. Elizabeth's Medical Center, Tufts University School of Medicine, 736 Cambridge St., Boston, MA 02135, United States.
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Li Y, Hu X, Liu Y, Bao Y, An L. Nimodipine protects dopaminergic neurons against inflammation-mediated degeneration through inhibition of microglial activation. Neuropharmacology 2008; 56:580-9. [PMID: 19049811 DOI: 10.1016/j.neuropharm.2008.10.016] [Citation(s) in RCA: 72] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2008] [Revised: 10/13/2008] [Accepted: 10/28/2008] [Indexed: 01/09/2023]
Abstract
Nimodipine, a calcium channel blocker, has been used mainly in the therapy of cardiovascular diseases. Recently, its indications have been extended experimentally to a wider range of disorders especially some central nervous system (CNS) disorders. In this study, we investigated whether nimodipine is neuroprotective to inflammation-mediated neurodegenerative diseases. Pretreatment with nimodipine reduced the degeneration of dopaminergic (DA) neurons induced by LPS in mesencephalic neuron-glia cultures in a dose-dependent manner. The neuroprotective effect of nimodipine was attributed to the inhibition of microglial activation, since nimodipine significantly inhibited the production of nitric oxide (NO), tumor necrosis factor-alpha (TNF-alpha), interleukin-1beta (IL-1beta) and prostaglandin E(2) (PGE(2)) from LPS-stimulated microglia. Moreover, nimodipine was not neuroprotective to 1-methyi-4-phenylpyridinium (MPP(+))-induced DA neurotoxicity in the absence of microglia. Mechanistic study showed that nimodipine failed to protect the degeneration of neurons in neuron-glia cultures from mice lacking functional NADPH oxidase (PHOX), a key enzyme for extracellular superoxide production in immune cells. Taken together these results suggest that nimodipine is protective to DA neurodegeneration via inhibiting the microglial-mediated oxidative stress and inflammatory response. Thus, nimodipine may be a potential therapeutic agent for the treatment of inflammation-related neurodegenerative disorders such as Parkinson's disease.
Collapse
Affiliation(s)
- Yachen Li
- School of Environmental and Biological Science and Technology, Dalian University of Technology, No. 2 Linggong Road, Dalian, Liaoning 116024, China
| | | | | | | | | |
Collapse
|
46
|
Jang H, Zheng J, Lal R, Nussinov R. New structures help the modeling of toxic amyloidbeta ion channels. Trends Biochem Sci 2008; 33:91-100. [PMID: 18182298 DOI: 10.1016/j.tibs.2007.10.007] [Citation(s) in RCA: 123] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2007] [Revised: 10/18/2007] [Accepted: 10/19/2007] [Indexed: 11/16/2022]
Abstract
The mechanism of amyloid toxicity is poorly understood and there are two schools of thought in this hotly debated field: the first favors membrane destabilization by intermediate-to-large amyloid oligomers, with consequent thinning and non-specific ion leakage; the second favors ion-specific permeable channels lined by small amyloid oligomers. Published results currently support both mechanisms. However, the amyloidbeta (Abeta) peptide has recently been shown to form a U-shaped 'beta-strand-turn-beta-strand' structure. This structure and the available physiological data present a challenge for computational biology--to provide candidate models consistent with the experimental data. Modeling based on small Abeta oligomers containing extramembranous N-termini predicts channels with shapes and dimensions consistent with experimentally derived channel structures. These results support the hypothesis that small Abeta oligomers can form ion channels. Molecular dynamics modeling can provide blueprints of 3D structural conformations for many other amyloids whose membrane association is key to their toxicity.
Collapse
Affiliation(s)
- Hyunbum Jang
- Center for Cancer Research Nanobiology Program, SAIC-Frederick Inc. NCI-Frederick, Frederick, MD 21702, USA
| | | | | | | |
Collapse
|
47
|
Sultana R, Butterfield DA. Alterations of some membrane transport proteins in Alzheimer's disease: role of amyloid β-peptide. ACTA ACUST UNITED AC 2008; 4:36-41. [DOI: 10.1039/b715278g] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
|
48
|
Madrigal JLM, Kalinin S, Richardson JC, Feinstein DL. Neuroprotective actions of noradrenaline: effects on glutathione synthesis and activation of peroxisome proliferator activated receptor delta. J Neurochem 2007; 103:2092-101. [PMID: 17854349 DOI: 10.1111/j.1471-4159.2007.04888.x] [Citation(s) in RCA: 66] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The endogenous neurotransmitter noradrenaline (NA) can protect neurons from the toxic consequences of various inflammatory stimuli, however the exact mechanisms of neuroprotection are not well known. In the current study, we examined neuroprotective effects of NA in primary cultures of rat cortical neurons. Exposure to oligomeric amyloid beta (Abeta) 1-42 peptide induced neuronal damage revealed by increased staining with fluorojade, and toxicity assessed by LDH release. Abeta-dependent neuronal death did not involve neuronal expression of the inducible nitric oxide synthase 2 (NOS2), since Abeta did not induce nitrite production from neurons, LDH release was not reduced by co-incubation with NOS2 inhibitors, and neurotoxicity was similar in wildtype and NOS2 deficient neurons. Co-incubation with NA partially reduced Abeta-induced neuronal LDH release, and completely abrogated the increase in fluorojade staining. Treatment of neurons with NA increased expression of gamma-glutamylcysteine ligase, reduced levels of GSH peroxidase, and increased neuronal GSH levels. The neuroprotective effects of NA were partially blocked by co-treatment with an antagonist of peroxisome proliferator activated receptors (PPARs), and replicated by incubation with a selective PPARdelta (PPARdelta) agonist. NA also increased expression and activation of PPARdelta. Together these data demonstrate that NA can protect neurons from Abeta-induced damage, and suggest that its actions may involve activation of PPARdelta and increases in GSH production.
Collapse
Affiliation(s)
- Jose L M Madrigal
- Department of Anesthesiology, University of Illinois & Jesse Brown Veteran's Affairs Hospital, Chicago, Illinois, USA
| | | | | | | |
Collapse
|
49
|
Lopez JR, Lyckman A, Oddo S, Laferla FM, Querfurth HW, Shtifman A. Increased intraneuronal resting [Ca2+] in adult Alzheimer's disease mice. J Neurochem 2007; 105:262-71. [PMID: 18021291 DOI: 10.1111/j.1471-4159.2007.05135.x] [Citation(s) in RCA: 133] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Neurodegeneration in Alzheimer's disease (AD) has been linked to intracellular accumulation of misfolded proteins and dysregulation of intracellular Ca2+. In the current work, we determined the contribution of specific Ca2+ pathways to an alteration in Ca2+ homeostasis in primary cortical neurons from an adult triple transgenic (3xTg-AD) mouse model of AD that exhibits intraneuronal accumulation of beta-amyloid proteins. Resting free Ca2+ concentration ([Ca2+](i)), as measured with Ca2+-selective microelectrodes, was greatly elevated in neurons from 3xTg-AD and APP(SWE) mouse strains when compared with their respective non-transgenic neurons, while there was no alteration in the resting membrane potential. In the absence of the extracellular Ca2+, the [Ca2+](i) returned to near normal levels in 3xTg-AD neurons, demonstrating that extracellular Ca2+contributed to elevated [Ca2+](i). Application of nifedipine, or a non-L-type channel blocker, SKF-96365, partially reduced [Ca2+](i). Blocking the ryanodine receptors, with ryanodine or FLA-365 had no effect, suggesting that these channels do not contribute to the elevated [Ca2+](i). Conversely, inhibition of inositol trisphosphate receptors with xestospongin C produced a partial reduction in [Ca2+](i). These results demonstrate that an elevation in resting [Ca2+](i), contributed by aberrant Ca2+entry and release pathways, should be considered a major component of the abnormal Ca2+ homeostasis associated with AD.
Collapse
Affiliation(s)
- José R Lopez
- Department of Anesthesia, Brigham & Women's Hospital, Harvard Medical School, Boston, Massachusetts, USA
| | | | | | | | | | | |
Collapse
|
50
|
Thibault O, Gant JC, Landfield PW. Expansion of the calcium hypothesis of brain aging and Alzheimer's disease: minding the store. Aging Cell 2007; 6:307-17. [PMID: 17465978 PMCID: PMC1974776 DOI: 10.1111/j.1474-9726.2007.00295.x] [Citation(s) in RCA: 292] [Impact Index Per Article: 16.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Evidence accumulated over more than two decades has implicated Ca2+ dysregulation in brain aging and Alzheimer's disease (AD), giving rise to the Ca2+ hypothesis of brain aging and dementia. Electrophysiological, imaging, and behavioral studies in hippocampal or cortical neurons of rodents and rabbits have revealed aging-related increases in the slow afterhyperpolarization, Ca2+ spikes and currents, Ca2+ transients, and L-type voltage-gated Ca2+ channel (L-VGCC) activity. Several of these changes have been associated with age-related deficits in learning or memory. Consequently, one version of the Ca2+ hypothesis has been that increased L-VGCC activity drives many of the other Ca2+-related biomarkers of hippocampal aging. In addition, other studies have reported aging- or AD model-related alterations in Ca2+ release from ryanodine receptors (RyR) on intracellular stores. The Ca2+-sensitive RyR channels amplify plasmalemmal Ca2+ influx by the mechanism of Ca2+-induced Ca2+ release (CICR). Considerable evidence indicates that a preferred functional link is present between L-VGCCs and RyRs which operate in series in heart and some brain cells. Here, we review studies implicating RyRs in altered Ca2+ regulation in cell toxicity, aging, and AD. A recent study from our laboratory showed that increased CICR plays a necessary role in the emergence of Ca2+-related biomarkers of aging. Consequently, we propose an expanded L-VGCC/Ca2+ hypothesis, in which aging/pathological changes occur in both L-type Ca2+ channels and RyRs, and interact to abnormally amplify Ca2+ transients. In turn, the increased transients result in dysregulation of multiple Ca2+-dependent processes and, through somewhat different pathways, in accelerated functional decline during aging and AD.
Collapse
Affiliation(s)
- Olivier Thibault
- Department of Molecular and Biomedical Pharmacology, University of Kentucky, University of Kentucky Medical Center, Lexington, KY 40536, USA
| | | | | |
Collapse
|