1
|
Prasanth MI, Verma K, Brimson S, Tencomnao T, Brimson JM. Simple ammonium salt and sigma-1 receptor ligand dipentylammonium provides neuroprotective effects in cell culture and Caenorhabditis elegans models of Alzheimer's disease. Biomed Pharmacother 2024; 173:116455. [PMID: 38503234 DOI: 10.1016/j.biopha.2024.116455] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2024] [Revised: 03/08/2024] [Accepted: 03/15/2024] [Indexed: 03/21/2024] Open
Abstract
The sigma-1 receptor (σ-1R), a chaperone protein located at the mitochondria-associated membrane (MAM) of the endoplasmic reticulum, can interact with and modify the signaling pathways of various proteins, thereby modulating many disease pathologies, including Alzheimer's disease (AD). The σ-1R ligand dipentylammonium (DPA) was analyzed for its anti-AD properties using PC12 cells (in vitro) and Caenorhabditis elegans (in vivo) models along with molecular docking (in silico) analysis. DPA at 1 and 10 µM concentrations was able to significantly potentiate NGF-induced neurite growth length by 137.7 ± 12.0 and 187.8 ± 16.4, respectively, when compared to the control 76.9 ± 7.4. DPA also regulated neurite damage caused by Aβ(25-35) treatment in differentiated PC12 cells by improving cell viability and neurite length. In C. elegans, DPA could significantly extend the median and maximum lifespan of Aβ transgenic strain CL2006 without impacting wild-type nematodes. Additionally, it could significantly reduce the paralysis phenotype of another Aβ transgenic strain, CL4176, thereby improving the overall health in AD pathogenesis. This effect depended on σ-1R, as DPA could not modulate the lifespan of σ-1R mutant TM3443. This was further confirmed using agonist PRE084 and antagonist BD1047, wherein the agonist alone could extend the lifespan of CL2006, while the antagonist suppressed the effect of DPA in CL2006. Interestingly, neither had an TM3443. Further, molecular docking analysis showed that DPA had a similar binding affinity as that of PRE084, BD1047 and pentazocine against the σ-1R receptor in humans and C. elegans, which collectively suggests the anti-AD properties of DPA.
Collapse
Affiliation(s)
- Mani Iyer Prasanth
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Kanika Verma
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Department of Molecular Epidemiology, ICMR-National Institute of Malaria Research (NIMR), New Delhi 110077, India
| | - Sirikalaya Brimson
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| | - James Michael Brimson
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand; Research Unit for Innovation and International Affairs, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok 10330, Thailand.
| |
Collapse
|
2
|
Munguia-Galaviz FJ, Miranda-Diaz AG, Cardenas-Sosa MA, Echavarria R. Sigma-1 Receptor Signaling: In Search of New Therapeutic Alternatives for Cardiovascular and Renal Diseases. Int J Mol Sci 2023; 24:ijms24031997. [PMID: 36768323 PMCID: PMC9916216 DOI: 10.3390/ijms24031997] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/30/2022] [Revised: 01/13/2023] [Accepted: 01/17/2023] [Indexed: 01/20/2023] Open
Abstract
Cardiovascular and renal diseases are among the leading causes of death worldwide, and regardless of current efforts, there is a demanding need for therapeutic alternatives to reduce their progression to advanced stages. The stress caused by diseases leads to the activation of protective mechanisms in the cell, including chaperone proteins. The Sigma-1 receptor (Sig-1R) is a ligand-operated chaperone protein that modulates signal transduction during cellular stress processes. Sig-1R interacts with various ligands and proteins to elicit distinct cellular responses, thus, making it a potential target for pharmacological modulation. Furthermore, Sig-1R ligands activate signaling pathways that promote cardioprotection, ameliorate ischemic injury, and drive myofibroblast activation and fibrosis. The role of Sig-1R in diseases has also made it a point of interest in developing clinical trials for pain, neurodegeneration, ischemic stroke, depression in patients with heart failure, and COVID-19. Sig-1R ligands in preclinical models have significantly beneficial effects associated with improved cardiac function, ventricular remodeling, hypertrophy reduction, and, in the kidney, reduced ischemic damage. These basic discoveries could inform clinical trials for heart failure (HF), myocardial hypertrophy, acute kidney injury (AKI), and chronic kidney disease (CKD). Here, we review Sig-1R signaling pathways and the evidence of Sig-1R modulation in preclinical cardiac and renal injury models to support the potential therapeutic use of Sig-1R agonists and antagonists in these diseases.
Collapse
Affiliation(s)
- Francisco Javier Munguia-Galaviz
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
- Division de Ciencias de la Salud, Centro Universitario del Sur, Universidad de Guadalajara, Ciudad Guzman 49000, Jalisco, Mexico
| | - Alejandra Guillermina Miranda-Diaz
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Miguel Alejandro Cardenas-Sosa
- Departamento de Fisiologia, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara 44340, Jalisco, Mexico
| | - Raquel Echavarria
- CONACYT-Centro de Investigacion Biomedica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara 44340, Jalisco, Mexico
- Correspondence:
| |
Collapse
|
3
|
Kabra A, Garg R, Brimson J, Živković J, Almawash S, Ayaz M, Nawaz A, Hassan SSU, Bungau S. Mechanistic insights into the role of plant polyphenols and their nano-formulations in the management of depression. Front Pharmacol 2022; 13:1046599. [PMID: 36419621 PMCID: PMC9676275 DOI: 10.3389/fphar.2022.1046599] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2022] [Accepted: 10/24/2022] [Indexed: 01/07/2024] Open
Abstract
Depression is a condition characterized by low mood and an aversion to activity, that causes behavioral problems, poor quality of life and limits daily life activities. It is considered as the fourth leading cause of disability worldwide. Selective Serotonin Reuptake Inhibitors (SSRIs) Monoamine Oxidase (MAO) inhibitors, Tricyclic Antidepressants (TCAs), and atypical antidepressants are some of the conventional medications used to treat depression. However, only about half of patients with major depressive disorder (MDD) respond effectively to first-line antidepressant therapy. Additionally, there are a number of drawbacks to standard antidepressants, such as anti-cholinergic side effects, drug-drug interactions, and food-drug interactions, which prompts researchers to look at alternative approaches to the treatment of depression. Medicinal plants and their metabolites are extensively tested for their efficacy against depression. Electronic databases such as Google scholar, Science Direct, SciFinder and PubMed were used to search relevant literature on the role of polyphenols in depression. Plants-derived Polyphenols represent a major class of compounds extensively distributed in plants. Number of polyphenols have demonstrated antidepressant activity, among which berberine, piperine, curcumin, naringenin, ascorbic acid and ginsenosides are extensively evaluated. The medicinal plants and their derived compounds mediated synthesized green nanoparticles have also exhibited considerable efficacy in the management of depression. The therapeutic effects of these phytochemicals is mediated via differentiation and inhibition of neuronal cell apoptosis, promotion of neuronal cell survival and modulation of key neurotransmitters. The aim of this study is to review compressively the chemical, pharmacological and neurological evidence showing the potential of polyphenols in depression.
Collapse
Affiliation(s)
- Atul Kabra
- University Institute of Pharma Sciences, Chandigarh University, Mohali, Punjab, India
| | - Ruchika Garg
- University School of Pharmaceutical Sciences, Rayat Bhara University, Mohali, Punjab, India
| | - James Brimson
- Natural Products for Neuroprotection and Anti-Ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
| | - Jelena Živković
- Department for Pharmaceutical Research and Development, Institute for Medicinal Plants Research “Dr. Josif Pančić”, Belgrade, Serbia
| | - Saud Almawash
- Department of Pharmaceutical Sciences, College of Pharmacy, Shaqra University, Shaqra, Saudi Arabia
| | - Muhammad Ayaz
- Department of Pharmacy, Faculty of Biological Sciences, University of Malakand, Chakdara, Pakistan
| | - Asif Nawaz
- Department of Pharmacy, Faculty of Biological Sciences, University of Malakand, Chakdara, Pakistan
| | - Syed Shams Ul Hassan
- Shanghai Key Laboratory for Molecular Engineering of Chiral Drugs, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
- Department of Natural Product Chemistry, School of Pharmacy, Shanghai Jiao Tong University, Shanghai, China
| | - Simona Bungau
- Department of Pharmacy, Faculty of Medicine and Pharmacy, University of Oradea, Oradea, Romania
| |
Collapse
|
4
|
Wang YM, Xia CY, Jia HM, He J, Lian WW, Yan Y, Wang WP, Zhang WK, Xu JK. Sigma-1 receptor: A potential target for the development of antidepressants. Neurochem Int 2022; 159:105390. [PMID: 35810915 DOI: 10.1016/j.neuint.2022.105390] [Citation(s) in RCA: 7] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2022] [Revised: 06/10/2022] [Accepted: 07/05/2022] [Indexed: 10/17/2022]
Abstract
Though a great many of studies on the development of antidepressants for the therapy of major depression disorder (MDD) and the development of antidepressants have been carried out, there still lacks an efficient approach in clinical practice. The involvement of Sigma-1 receptor in the pathological process of MDD has been verified. In this review, recent research focusing on the role of Sigma-1 receptor in the etiology of MDD were summarized. Preclinical studies and clinical trials have found that stress induce the variation of Sigma-1 receptor in the blood, brain and heart. Dysfunction and absence of Sigma-1 receptor result in depressive-like behaviors in rodent animals. Agonists of Sigma-1 receptor show not only antidepressant-like activities but also therapeutical effects in complications of depression. The mechanisms underlying antidepressant-like effects of Sigma-1 receptor may include suppressing neuroinflammation, regulating neurotransmitters, ameliorating brain-derived neurotrophic factor and N-Methyl-D-Aspartate receptor, and alleviating the endoplasmic reticulum stress and mitochondria damage during stress. Therefore, Sigma-1 receptor represents a potential target for antidepressants development.
Collapse
Affiliation(s)
- Yu-Ming Wang
- School of Life Sciences & School of Chinese Medicine Sciences, Beijing University of Chinese Medicine, Beijing, 100029, PR China; Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Cong-Yuan Xia
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Hong-Mei Jia
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing, PR China
| | - Jun He
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Wen-Wen Lian
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Yu Yan
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Wen-Ping Wang
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China
| | - Wei-Ku Zhang
- Institute of Clinical Medical Sciences & Department of Pharmacy, China-Japan Friendship Hospital, Beijing, 100029, PR China.
| | - Jie-Kun Xu
- School of Life Sciences & School of Chinese Medicine Sciences, Beijing University of Chinese Medicine, Beijing, 100029, PR China.
| |
Collapse
|
5
|
Wei X, Zheng Z, Feng Z, Zheng L, Tao S, Zheng B, Huang B, Zhang X, Liu J, Chen Y, Zong W, Shan Z, Fan S, Chen J, Zhao F. Sigma-1 receptor attenuates osteoclastogenesis by promoting ER-associated degradation of SERCA2. EMBO Mol Med 2022; 14:e15373. [PMID: 35611810 PMCID: PMC9260208 DOI: 10.15252/emmm.202115373] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 05/10/2022] [Accepted: 05/11/2022] [Indexed: 12/04/2022] Open
Abstract
Sigma-1 receptor (Sigmar1) is a specific chaperone located in the mitochondria-associated endoplasmic reticulum membrane (MAM) and plays a role in several physiological processes. However, the role of Sigmar1 in bone homeostasis remains unknown. Here, we show that mice lacking Sigmar1 exhibited severe osteoporosis in an ovariectomized model. In contrast, overexpression of Sigmar1 locally alleviated the osteoporosis phenotype. Treatment with Sigmar1 agonists impaired both human and mice osteoclast formation in vitro. Mechanistically, SERCA2 was identified to interact with Sigmar1 based on the immunoprecipitation-mass spectrum (IP-MS) and co-immunoprecipitation (co-IP) assays, and Q615 of SERCA2 was confirmed to be the critical residue for their binding. Furthermore, Sigmar1 promoted SERCA2 degradation through Hrd1/Sel1L-dependent ER-associated degradation (ERAD). Ubiquitination of SERCA2 at K460 and K541 was responsible for its proteasomal degradation. Consequently, inhibition of SERCA2 impeded Sigmar1 deficiency enhanced osteoclastogenesis. Moreover, we found that dimemorfan, an FDA-approved Sigmar1 agonist, effectively rescued bone mass in various established bone-loss models. In conclusion, Sigmar1 is a negative regulator of osteoclastogenesis, and activation of Sigmar1 by dimemorfan may be a potential treatment for osteoporosis in clinical practice.
Collapse
Affiliation(s)
- Xiaoan Wei
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and RegenerationTranslational Research of Zhejiang ProvinceHangzhouChina
| | - Zeyu Zheng
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and RegenerationTranslational Research of Zhejiang ProvinceHangzhouChina
| | - Zhenhua Feng
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and RegenerationTranslational Research of Zhejiang ProvinceHangzhouChina
| | - Lin Zheng
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and RegenerationTranslational Research of Zhejiang ProvinceHangzhouChina
| | - Siyue Tao
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and RegenerationTranslational Research of Zhejiang ProvinceHangzhouChina
| | - Bingjie Zheng
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and RegenerationTranslational Research of Zhejiang ProvinceHangzhouChina
| | - Bao Huang
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and RegenerationTranslational Research of Zhejiang ProvinceHangzhouChina
| | - Xuyang Zhang
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and RegenerationTranslational Research of Zhejiang ProvinceHangzhouChina
| | - Junhui Liu
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and RegenerationTranslational Research of Zhejiang ProvinceHangzhouChina
| | - Yilei Chen
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and RegenerationTranslational Research of Zhejiang ProvinceHangzhouChina
| | - Wentian Zong
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and RegenerationTranslational Research of Zhejiang ProvinceHangzhouChina
| | - Zhi Shan
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and RegenerationTranslational Research of Zhejiang ProvinceHangzhouChina
| | - Shunwu Fan
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and RegenerationTranslational Research of Zhejiang ProvinceHangzhouChina
| | - Jian Chen
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and RegenerationTranslational Research of Zhejiang ProvinceHangzhouChina
| | - Fengdong Zhao
- Department of Orthopaedic SurgerySir Run Run Shaw HospitalZhejiang University School of MedicineHangzhouChina
- Key Laboratory of Musculoskeletal System Degeneration and RegenerationTranslational Research of Zhejiang ProvinceHangzhouChina
| |
Collapse
|
6
|
Sun Y, Wan W, Zhao X, Han X, Ye T, Chen X, Ran Q, Wang X, Liu X, Qu C, Shi S, Zhang C, Yang B. Chronic Sigma 1 receptor activation alleviates right ventricular dysfunction secondary to pulmonary arterial hypertension. Bioengineered 2022; 13:10843-10856. [PMID: 35473584 PMCID: PMC9208487 DOI: 10.1080/21655979.2022.2065953] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Sigma 1 receptor (S1R) has shown a preferable protective effect on left ventricular function, but whether it protects right ventricular (RV) function is still elusive.This study aimed to determine the effects of S1R on RV dysfunction secondary to pulmonary arterial hypertension.Sixty wild-type male Sprague–Dawley rats were randomly divided into the control group, the fluvoxamine group, the pulmonary arterial hypertension group and the pulmonary arterial hypertension combined with fluvoxamine group. Monocrotaline (60 mg/kg) was administered to induce pulmonary arterial hypertension, and fluvoxamine was given for 21 consecutive days to activate S1R after one week of monocrotaline administration. Echocardiographic, serologic, and histologic parameters, qRT-PCR, and western blotting were conducted after 4 weeks of monocrotaline administration.The expression of S1R was decreased in the right ventricle in pulmonary arterial hypertension. TAPSE, and the FAC of the right ventricle were significantly decreased, and RV EDP and the plasma concentration of N-terminal pro-B-type natriuretic peptide was increased in the pulmonary arterial hypertension group, but fluvoxamine partly restored those abnormalities (all P < 0.05). Moreover, pulmonary arteriole remodeling, and fibrosis and hypertrophy in the RV were shown in the pulmonary arterial hypertension group; interestingly, fluvoxamine recovered RV structural remodeling (all P < 0.05) but neither alleviated pulmonary arteriole remodeling nor reduced pulmonary artery pressure. Furthermore, S1R activation protects RV function by upgrading the NRF 2/HO 1-mediated antioxidant stress pathway. In conclusion, chronic S1R activation ameliorates structural remodeling and RV dysfunction secondary to pulmonary arterial hypertension without altering pulmonary artery pressure.
Collapse
Affiliation(s)
- Yazhou Sun
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Weiguo Wan
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xin Zhao
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xueyu Han
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Tianxin Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xiaoli Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Qian Ran
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xiukun Wang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Shaobo Shi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Cui Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, Hubei, China.,Cardiovascular Research Institute, Wuhan University, Wuhan, Hubei, China.,Hubei Key Laboratory of Cardiology, Wuhan, Hubei, China
| |
Collapse
|
7
|
Sałaciak K, Pytka K. Revisiting the sigma-1 receptor as a biological target to treat affective and cognitive disorders. Neurosci Biobehav Rev 2022; 132:1114-1136. [PMID: 34736882 PMCID: PMC8559442 DOI: 10.1016/j.neubiorev.2021.10.037] [Citation(s) in RCA: 19] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 10/25/2021] [Accepted: 10/28/2021] [Indexed: 12/21/2022]
Abstract
Depression and cognitive disorders are diseases with complex and not-fully understood etiology. Unfortunately, the COVID-19 pandemic dramatically increased the prevalence of both conditions. Since the current treatments are inadequate in many patients, there is a constant need for discovering new compounds, which will be more effective in ameliorating depressive symptoms and treating cognitive decline. Proteins attracting much attention as potential targets for drugs treating these conditions are sigma-1 receptors. Sigma-1 receptors are multi-functional proteins localized in endoplasmic reticulum membranes, which play a crucial role in cellular signal transduction by interacting with receptors, ion channels, lipids, and kinases. Changes in their functions and expression may lead to various diseases, including depression or memory impairments. Thus, sigma-1 receptor modulation might be useful in treating these central nervous system diseases. Importantly, two sigma-1 receptor ligands entered clinical trials, showing that this compound group possesses therapeutic potential. Therefore, based on preclinical studies, this review discusses whether the sigma-1 receptor could be a promising target for drugs treating affective and cognitive disorders.
Collapse
Affiliation(s)
- Kinga Sałaciak
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland
| | - Karolina Pytka
- Department of Pharmacodynamics, Faculty of Pharmacy, Jagiellonian University Medical College, Medyczna 9, 30-688 Krakow, Poland.
| |
Collapse
|
8
|
Ye Y, Zhou CC, Hu HQ, Fukuzawa I, Zhang HL. Underlying mechanisms of acupuncture therapy on polycystic ovary syndrome: Evidences from animal and clinical studies. Front Endocrinol (Lausanne) 2022; 13:1035929. [PMID: 36353235 PMCID: PMC9637827 DOI: 10.3389/fendo.2022.1035929] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/03/2022] [Accepted: 10/11/2022] [Indexed: 11/13/2022] Open
Abstract
Polycystic ovary syndrome (PCOS) is a common endocrine and metabolic disorder among women of reproductive age. Current standard treatment includes lifestyle change, oral pharmacological agents, and surgical modalities. However, the efficacy of current therapies is less than satisfactory. Clinical evidence has shown that acupuncture is effective for regulating hormone levels, promoting ovulation, and attenuating insulin resistance in patients with PCOS. Acupuncture may affect the production of β-endorphin, which may lead to gonadotropin-releasing hormone secretion and then affect ovulation, menstrual cycle, and fertility. The mechanism of acupuncture for patients with PCOS has not been comprehensively reviewed so far. Better understanding of the mechanisms of acupuncture would help popularize the use of acupuncture therapy for patients with PCOS. In this narrative review, we aimed to overview the potential mechanisms and evidence-based data of acupuncture on PCOS, and analyze the most frequently used acupoints based on animal and clinical studies. The results of this study will contribute to a better understanding of the current situation in this field.
Collapse
Affiliation(s)
- Yang Ye
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Cong-Cong Zhou
- School of Global Public Health, New York University, New York, NY, United States
| | - Hang-Qi Hu
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Ii Fukuzawa
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
| | - Hao-Lin Zhang
- Department of Traditional Chinese Medicine, Peking University Third Hospital, Beijing, China
- *Correspondence: Hao-Lin Zhang,
| |
Collapse
|
9
|
Aishwarya R, Abdullah CS, Morshed M, Remex NS, Bhuiyan MS. Sigmar1's Molecular, Cellular, and Biological Functions in Regulating Cellular Pathophysiology. Front Physiol 2021; 12:705575. [PMID: 34305655 PMCID: PMC8293995 DOI: 10.3389/fphys.2021.705575] [Citation(s) in RCA: 34] [Impact Index Per Article: 11.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2021] [Accepted: 06/07/2021] [Indexed: 12/11/2022] Open
Abstract
The Sigma 1 receptor (Sigmar1) is a ubiquitously expressed multifunctional inter-organelle signaling chaperone protein playing a diverse role in cellular survival. Recessive mutation in Sigmar1 have been identified as a causative gene for neuronal and neuromuscular disorder. Since the discovery over 40 years ago, Sigmar1 has been shown to contribute to numerous cellular functions, including ion channel regulation, protein quality control, endoplasmic reticulum-mitochondrial communication, lipid metabolism, mitochondrial function, autophagy activation, and involved in cellular survival. Alterations in Sigmar1’s subcellular localization, expression, and signaling has been implicated in the progression of a wide range of diseases, such as neurodegenerative diseases, ischemic brain injury, cardiovascular diseases, diabetic retinopathy, cancer, and drug addiction. The goal of this review is to summarize the current knowledge of Sigmar1 biology focusing the recent discoveries on Sigmar1’s molecular, cellular, pathophysiological, and biological functions.
Collapse
Affiliation(s)
- Richa Aishwarya
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Chowdhury S Abdullah
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Mahboob Morshed
- Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Naznin Sultana Remex
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| | - Md Shenuarin Bhuiyan
- Department of Molecular and Cellular Physiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States.,Department of Pathology and Translational Pathobiology, Louisiana State University Health Sciences Center-Shreveport, Shreveport, LA, United States
| |
Collapse
|
10
|
Brimson JM, Prasanth MI, Malar DS, Brimson S, Thitilertdecha P, Tencomnao T. Drugs that offer the potential to reduce hospitalization and mortality from SARS-CoV-2 infection: The possible role of the sigma-1 receptor and autophagy. Expert Opin Ther Targets 2021; 25:435-449. [PMID: 34236922 PMCID: PMC8290373 DOI: 10.1080/14728222.2021.1952987] [Citation(s) in RCA: 23] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/02/2021] [Accepted: 07/05/2021] [Indexed: 02/07/2023]
Abstract
Introduction: Despite the availability of new vaccines for SARS-CoV-2, there has been slow uptake and problems with supply in some parts of the world. Hence, there is still a necessity for drugs that can prevent hospitalization of patients and reduce the strain on health care systems. Drugs with sigma affinity potentially provide protection against the most severe symptoms of SARS-COV-2 and could prevent mortality via interactions with the sigma-1 receptor.Areas covered: This review examines the role of the sigma-1 receptor and autophagy in SARS-CoV-2 infections and how they may be linked. The authors reveal how sigma ligands may reduce the symptoms, complications, and deaths resulting from SARS-CoV-2 and offer insights on those patient cohorts that may benefit most from these drugs.Expert opinion: Drugs with sigma affinity potentially offer protection against the most severe symptoms of SARS-CoV-2 via interactions with the sigma-1 receptor. Agonists of the sigma-1 receptor may provide protection of the mitochondria, activate mitophagy to remove damaged and leaking mitochondria, prevent ER stress, manage calcium ion transport, and induce autophagy to prevent cell death in response to infection.
Collapse
Affiliation(s)
- James Michael Brimson
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Mani Iyer Prasanth
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Dicson Sheeja Malar
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Sirikalaya Brimson
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| | - Premrutai Thitilertdecha
- Siriraj Research Group in Immunobiology and Therapeutic Sciences, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Tewin Tencomnao
- Natural Products for Neuroprotection and Anti-ageing Research Unit, Chulalongkorn University, Bangkok, Thailand
- Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University, Bangkok, Thailand
| |
Collapse
|
11
|
Guo Y, Zhang C, Chen X, Liu X, Ye T, Fo Y, Shi S, Qu C, Liang J, Shen B, Yang B. Sigma-1 receptor ligands improves ventricular repolarization-related ion remodeling in rats with major depression disorder. Psychopharmacology (Berl) 2021; 238:487-499. [PMID: 33140216 DOI: 10.1007/s00213-020-05697-4] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/27/2020] [Accepted: 10/23/2020] [Indexed: 02/07/2023]
Abstract
RATIONALE It has been reported that patients with major depressive disorder (MDD) are prone to developing ventricular arrhythmias. Moreover, the Sigma-1 receptor not only plays a crucial role in MDD but has also been shown to have antiarrhythmic properties. The Sigma-1 receptor is a common receptor related to depression and ventricular arrhythmias. OBJECTIVE We analyzed the effects of the Sigma-1 receptor on depression and ventricular repolarization-related ion remodeling in MDD rats. METHODS MDD was induced in rats by chronic unpredictable mild stress (CUMS), and 28 days later, the rats were subjected to behavior tests. Protein expression was measured by western blotting, and cardiac morphological changes were observed by Masson staining. Electrophysiological measurement of the myocardium was performed with the whole-cell patch-clamp technique. RESULTS Compared with the control rats, the MDD rats exhibited lower transient outward potassium current (Ito) and L-type calcium current (ICa-L) amplitudes. On the other hand, a trend of depolarization of Ito and hyperpolarization of ICa-L was observed in the MDD rats. Thus, we investigated the effect of fluvoxamine, a Sigma-1 receptor agonist, on Ito and ICa-L. Fluvoxamine enhanced Ito and altered its current kinetics, as shown by acceleration of activation and recovery from inactivation. In contrast, fluvoxamine inhibited the Ca2+ by hyperpolarizing the steady-state activation of ICa-L. All these effects were blocked by BD1047. CONCLUSION Taken together, our results indicate that Sigma-1 receptor modulates the functions of Ito and ICa-L to possibly exert antiarrhythmic effects.
Collapse
Affiliation(s)
- Yan Guo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Cui Zhang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Xiuhuan Chen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Xin Liu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Tianxin Ye
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Yuhong Fo
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Shaobo Shi
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Chuan Qu
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Jinjun Liang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China
| | - Bo Shen
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China.
| | - Bo Yang
- Department of Cardiology, Renmin Hospital of Wuhan University, Wuhan, People's Republic of China.
- Cardiovascular Research Institute, Wuhan University, Wuhan, People's Republic of China.
- Hubei Key Laboratory of Cardiology, Wuhan, People's Republic of China.
| |
Collapse
|
12
|
Voronin MV, Vakhitova YV, Seredenin SB. Chaperone Sigma1R and Antidepressant Effect. Int J Mol Sci 2020; 21:E7088. [PMID: 32992988 PMCID: PMC7582751 DOI: 10.3390/ijms21197088] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2020] [Revised: 09/17/2020] [Accepted: 09/22/2020] [Indexed: 12/13/2022] Open
Abstract
This review analyzes the current scientific literature on the role of the Sigma1R chaperone in the pathogenesis of depressive disorders and pharmacodynamics of antidepressants. As a result of ligand activation, Sigma1R is capable of intracellular translocation from the endoplasmic reticulum (ER) into the region of nuclear and cellular membranes, where it interacts with resident proteins. This unique property of Sigma1R provides regulation of various receptors, ion channels, enzymes, and transcriptional factors. The current review demonstrates the contribution of the Sigma1R chaperone to the regulation of molecular mechanisms involved in the antidepressant effect.
Collapse
Affiliation(s)
- Mikhail V. Voronin
- Department of Pharmacogenetics, FSBI “Zakusov Institute of Pharmacology”, Baltiyskaya Street 8, 125315 Moscow, Russia;
| | | | - Sergei B. Seredenin
- Department of Pharmacogenetics, FSBI “Zakusov Institute of Pharmacology”, Baltiyskaya Street 8, 125315 Moscow, Russia;
| |
Collapse
|
13
|
Brimson JM, Brimson S, Chomchoei C, Tencomnao T. Using sigma-ligands as part of a multi-receptor approach to target diseases of the brain. Expert Opin Ther Targets 2020; 24:1009-1028. [PMID: 32746649 DOI: 10.1080/14728222.2020.1805435] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
INTRODUCTION The sigma receptors are found abundantly in the central nervous system and are targets for the treatment of various diseases, including Alzheimer's (AD), Parkinson's (PD), Huntington's disease (HD), depression, amyotrophic lateral sclerosis (ALS), and multiple sclerosis (MS). However, for many of these diseases, other receptors and targets have been the focus of the most, such as acetylcholine esterase inhibitors in Alzheimer's and dopamine replacement in Parkinson's. The currently available drugs for these diseases have limited success resulting in the requirement of an alternative approach to their treatment. AREAS COVERED In this review, we discuss the potential role of the sigma receptors and their ligands as part of a multi receptor approach in the treatment of the diseases mentioned above. The literature reviewed was obtained through searches in databases, including PubMed, Web of Science, Google Scholar, and Scopus. EXPERT OPINION Given sigma receptor agonists provide neuroprotection along with other benefits such as potentiating the effects of other receptors, further development of multi-receptor targeting ligands, and or the development of multi-drug combinations to target multiple receptors may prove beneficial in the future treatment of degenerative diseases of the CNS, especially when coupled with better diagnostic techniques.
Collapse
Affiliation(s)
- James Michael Brimson
- Age-related Inflammation and Degeneration Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University , Bangkok, Thailand
| | - Sirikalaya Brimson
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University , Bangkok, Thailand
| | - Chanichon Chomchoei
- Department of Clinical Microscopy, Faculty of Allied Health Sciences, Chulalongkorn University , Bangkok, Thailand
| | - Tewin Tencomnao
- Age-related Inflammation and Degeneration Research Unit, Department of Clinical Chemistry, Faculty of Allied Health Sciences, Chulalongkorn University , Bangkok, Thailand
| |
Collapse
|
14
|
Mocayar Marón FJ, Ferder L, Saraví FD, Manucha W. Hypertension linked to allostatic load: from psychosocial stress to inflammation and mitochondrial dysfunction. Stress 2019; 22:169-181. [PMID: 30547701 DOI: 10.1080/10253890.2018.1542683] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/09/2023] Open
Abstract
Although a large number of available treatments and strategies, the prevalence of cardiovascular diseases continues to grow worldwide. Emerging evidence supports the notion of counteracting stress as a critical component of a comprehensive therapeutic strategy for cardiovascular disease. Indeed, an unhealthy lifestyle is a burden to biological variables such as plasma glucose, lipid profile, and blood pressure control. Recent findings identify allostatic load as a new paradigm for an integrated understanding of the importance of psychosocial stress and its impact on the development and maintenance of cardiovascular disease. Allostasis complement homeostasis and integrates behavioral and physiological mechanisms by which genes, early experiences, environment, lifestyle, diet, sleep, and physical exercise can modulate and adapt biological responses at the cellular level. For example, variability is a physiological characteristic of blood pressure necessary for survival and the allostatic load in hypertension can contribute to its related cardiovascular morbidity and mortality. Therefore, the current review will focus on the mechanisms that link hypertension to allostatic load, which includes psychosocial stress, inflammation, and mitochondrial dysfunction. We will describe and discuss new insights on neuroendocrine-immune effects linked to allostatic load and its impact on the cellular and molecular responses; the links between allostatic load, inflammation, and endothelial dysfunction; the epidemiological evidence supporting the pathophysiological origins of hypertension; and the biological embedding of allostatic load and hypertension with an emphasis on mitochondrial dysfunction.
Collapse
Affiliation(s)
- Feres José Mocayar Marón
- a Área de Química Biológica, Departamento de Morfofisiología, Facultad de Ciencias Médicas , Universidad Nacional de Cuyo , Mendoza , Argentina
| | - León Ferder
- b Department of Pediatrics , Nephrology Division, Miller School of Medicine, University of Miami , FL , USA
| | - Fernando Daniel Saraví
- c Instituto de Fisiología, Departamento de Morfofisiología, Facultad de Ciencias Médicas , Universidad Nacional de Cuyo , Mendoza , Argentina
| | - Walter Manucha
- d Área de Farmacología, Departamento de Patología, Facultad de Ciencias Médicas , Universidad Nacional de Cuyo , Mendoza , Argentina
- e Instituto de Medicina y Biología Experimental de Cuyo (IMBECU), Consejo Nacional de Investigaciones Científicas y Tecnológicas (CONICET) , Mendoza , Argentina
| |
Collapse
|
15
|
Abstract
More than four decades passed since sigma receptors were first mentioned. Since then, existence of at least two receptor subtypes and their tissue distributions have been proposed. Nowadays, it is clear, that sigma receptors are unique ubiquitous proteins with pluripotent function, which can interact with so many different classes of proteins. As the endoplasmic resident proteins, they work as molecular chaperones - accompany various proteins during their folding, ensure trafficking of the maturated proteins between cellular organelles and regulate their functions. In the heart, sigma receptor type 1 is more dominant. Cardiac sigma 1 receptors regulate response to endoplasmic reticulum stress, modulates calcium signaling in cardiomyocyte and can affect function of voltage-gated ion channels. They contributed in pathophysiology of cardiac hypertrophy, heart failure and many other cardiovascular disorders. Therefore, sigma receptors are potential novel targets for specific treatment of cardiovascular diseases.
Collapse
Affiliation(s)
- T Stracina
- Department of Physiology, Faculty of Medicine, Masaryk University, Brno, Czech Republic.
| | | |
Collapse
|
16
|
Imaging sigma receptors in the brain: New opportunities for diagnosis of Alzheimer's disease and therapeutic development. Neurosci Lett 2018; 691:3-10. [PMID: 30040970 DOI: 10.1016/j.neulet.2018.07.033] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2018] [Revised: 07/09/2018] [Accepted: 07/20/2018] [Indexed: 10/28/2022]
Abstract
The sigma-1 (σ1) receptor is a chaperone protein located on the mitochondria-associated membrane of the endoplasmic reticulum, while the sigma-2 receptor (σ2) is an endoplasmic reticulum-resident membrane protein. Recent evidence indicates that both of these receptors figure prominently in the pathophysiology of Alzheimer's disease (AD) and thus are targets for the development of novel, disease-modifying therapeutic strategies. Radioligand-based molecular imaging technique such as positron emission tomography (PET) imaging is a powerful tool for the investigation of protein target expression and function in living subjects. In this review, we survey the development of PET radioligands for the σ1 or σ2 receptors and assess their potential for human imaging applications. The availability of PET imaging with σ1 or σ2 receptor-specific radioligands in humans will allow the investigation of these receptors in vivo and lead to further understanding of their respective roles in AD pathogenesis and progression. Moreover, PET imaging can be used in target occupancy studies to assess target engagement and correlate receptor occupancy and therapeutic response of σ1 receptor agonists and σ2 receptor antagonists currently in clinical trials. It is expected that neuroimaging of σ1 and σ2 receptors in the brain will shed new light on AD pathophysiology and may provide us with new biomarkers for diagnosis of AD and efficacy monitoring of emerging AD therapeutic strategies.
Collapse
|
17
|
Abstract
Heart failure (HF) is a systemic illness with grave implications for bodily functions. The brain, among other vital organs, often suffers insults as a result of HF, and both anatomic and functional brain abnormalities were found in the HF population. This injury was demonstrated across a wide range of clinical conditions and cardiac functions and was shown to affect patients' outcomes. Although reduced cardiac output and high burden of cardiovascular risk factors are the prevailing explanations for these findings, there are data showing the involvement of neurohormonal, nutritional, and inflammatory mechanisms in this complex process. Here, the authors review the suggested pathophysiology behind brain injury in HF, describe its effect on patients' outcomes, offer a diagnostic approach, and discuss possible therapeutic options.
Collapse
|
18
|
He Y, Xie F, Ye J, Deuther-Conrad W, Cui B, Wang L, Lu J, Steinbach J, Brust P, Huang Y, Lu J, Jia H. 1-(4-[ 18F]Fluorobenzyl)-4-[(tetrahydrofuran-2-yl)methyl]piperazine: A Novel Suitable Radioligand with Low Lipophilicity for Imaging σ 1 Receptors in the Brain. J Med Chem 2017; 60:4161-4172. [PMID: 28409931 DOI: 10.1021/acs.jmedchem.6b01723] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
We have designed and synthesized novel piperazine compounds with low lipophilicity as σ1 receptor ligands. 1-(4-Fluorobenzyl)-4-[(tetrahydrofuran-2-yl)methyl]piperazine (10) possessed a low nanomolar σ1 receptor affinity and a high selectivity toward the vesicular acetylcholine transporter (>2000-fold), σ2 receptors (52-fold), and adenosine A2A, adrenergic α2, cannabinoid CB1, dopamine D1, D2L, γ-aminobutyric acid A (GABAA), NMDA, melatonin MT1, MT2, and serotonin 5-HT1 receptors. The corresponding radiotracer [18F]10 demonstrated high brain uptake and extremely high brain-to-blood ratios in biodistribution studies in mice. Pretreatment with the selective σ1 receptor agonist SA4503 significantly reduced the level of accumulation of the radiotracer in the brain. No radiometabolite of [18F]10 was observed to enter the brain. Positron emission tomography and magnetic resonance imaging confirmed suitable kinetics and a high specific binding of [18F]10 to σ1 receptors in rat brain. Ex vivo autoradiography showed a reduced level of binding of [18F]10 in the cortex and hippocampus of the senescence-accelerated prone (SAMP8) compared to that of the senescence-accelerated resistant (SAMR1) mice, indicating the potential dysfunction of σ1 receptors in Alzheimer's disease.
Collapse
Affiliation(s)
- Yingfang He
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University , Beijing, China
| | - Fang Xie
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University , Beijing, China
| | - Jiajun Ye
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University , Beijing, China
| | - Winnie Deuther-Conrad
- Helmholtz-Zentrum Dresden-Rossendorf , Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
| | - Bixiao Cui
- Department of Nuclear Medicine, Xuanwu Hospital Capital Medical University , Beijing, China
| | - Liang Wang
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University , Beijing, China
| | - Jie Lu
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University , Beijing, China
| | - Jörg Steinbach
- Helmholtz-Zentrum Dresden-Rossendorf , Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
| | - Peter Brust
- Helmholtz-Zentrum Dresden-Rossendorf , Institute of Radiopharmaceutical Cancer Research, Department of Neuroradiopharmaceuticals, 04318 Leipzig, Germany
| | - Yiyun Huang
- Yale PET Center, Department of Radiology and Biomedical Imaging, Yale University School of Medicine , New Haven, Connecticut 06520-8048, United States
| | - Jie Lu
- Department of Nuclear Medicine, Xuanwu Hospital Capital Medical University , Beijing, China
| | - Hongmei Jia
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University , Beijing, China
| |
Collapse
|
19
|
Carnevali L, Montano N, Statello R, Sgoifo A. Rodent models of depression-cardiovascular comorbidity: Bridging the known to the new. Neurosci Biobehav Rev 2017; 76:144-153. [DOI: 10.1016/j.neubiorev.2016.11.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2016] [Revised: 10/27/2016] [Accepted: 11/09/2016] [Indexed: 12/22/2022]
|
20
|
Fukunaga K, Moriguchi S. Stimulation of the Sigma-1 Receptor and the Effects on Neurogenesis and Depressive Behaviors in Mice. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 964:201-211. [PMID: 28315273 DOI: 10.1007/978-3-319-50174-1_14] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Sigma-1 receptor (Sig-1R) is molecular chaperone regulating calcium efflux from the neuronal endoplasmic reticulum to mitochondria. Recent studies show that Sig-1R stimulation antagonizes depressive-like behaviors in animal models, but molecular mechanisms underlying this effect remain unclear. Here, we focus on the effects of Sig-1R ligands on hippocampal neurogenesis and depressive-like behaviors. Sig-1R stimulation also enhances CaMKII /CaMKIV and protein kinase B (Akt) activities in hippocampus. Therefore, we discuss the fundamental roles of Sig-1R, CaMKII /CaMKIV and protein kinase B (Akt) signaling in amelioration of depressive-like behaviors following Sig-1R stimulation.
Collapse
Affiliation(s)
- Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi, 980-8578, Japan.
| | - Shigeki Moriguchi
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Miyagi, 980-8578, Japan
| |
Collapse
|
21
|
Shinoda Y, Tagashira H, Bhuiyan MS, Hasegawa H, Kanai H, Zhang C, Han F, Fukunaga K. Corticosteroids Mediate Heart Failure-Induced Depression through Reduced σ1-Receptor Expression. PLoS One 2016; 11:e0163992. [PMID: 27741227 PMCID: PMC5065174 DOI: 10.1371/journal.pone.0163992] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2016] [Accepted: 09/19/2016] [Indexed: 01/12/2023] Open
Abstract
Cardiovascular diseases are risk factors for depression in humans. We recently proposed that σ1 receptor (σ1R) stimulation rescued cardiac hypertrophy and heart failure induced by transverse aortic constriction (TAC) in mice. Importantly, σ1R stimulation reportedly ameliorates depression-like behaviors in rodents. Thus, we hypothesized that impaired σ1R activity in brain triggers depression-like behaviors in animals with cardiovascular disease. Indeed, here we found that cardiac hypertrophy and heart failure induced by TAC were associated with depression-like behaviors concomitant with downregulation of σ1R expression in brain 6 weeks after surgery. σ1R levels significantly decreased in astrocytes in both the hippocampal CA1 region and dentate gyrus. Oral administration of the specific σ1R agonist SA4503 (0.3-1.0mg/kg) significantly improved TAC-induced depression-like behaviors concomitant with rescued astrocytic σ1R expression in CA1 and the dentate gyrus. Plasma corticosterone levels significantly increased 6 weeks after TAC, and chronic treatment of mice with corticosterone for 3 weeks elicited depression-like behaviors concomitant with reduced astrocytic σ1R expression in hippocampus. Furthermore, the glucocorticoid receptor antagonist mifepristone antagonized depressive-like behaviors and ameliorated decreased hippocampal σ1R expression in TAC mice. We conclude that elevated corticosterone levels trigger hippocampal σ1R downregulation and that σ1R stimulation with SA4503 is an attractive therapy to improve not only cardiac dysfunction but depression-like behaviors associated with heart failure.
Collapse
Affiliation(s)
- Yasuharu Shinoda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Japan
| | - Hideaki Tagashira
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Japan
| | - Md. Shenuarin Bhuiyan
- Division of Molecular Cardiovascular Biology, Department of Pediatrics, Cincinnati Children’s Hospital Medical Center, Cincinnati, OH, 45229, United States of America
| | - Hideyuki Hasegawa
- Department of Electrical Engineering, Graduate School of Biomedical Engineering, Tohoku University, 6-6 Aramaki-Aoba, Aoba-ku, Sendai, Japan
| | - Hiroshi Kanai
- Department of Electrical Engineering, Graduate School of Biomedical Engineering, Tohoku University, 6-6 Aramaki-Aoba, Aoba-ku, Sendai, Japan
| | - Chen Zhang
- Department of Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 31005, P. R. China
| | - Feng Han
- Department of Pharmacy, College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, Zhejiang, 31005, P. R. China
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Japan
- * E-mail:
| |
Collapse
|
22
|
Ye J, Wang X, Deuther-Conrad W, Zhang J, Li J, Zhang X, Wang L, Steinbach J, Brust P, Jia H. Synthesis and evaluation of a18F-labeled 4-phenylpiperidine-4-carbonitrile radioligand for σ1receptor imaging. J Labelled Comp Radiopharm 2016; 59:332-9. [DOI: 10.1002/jlcr.3408] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2016] [Revised: 03/10/2016] [Accepted: 05/03/2016] [Indexed: 11/10/2022]
Affiliation(s)
- Jiajun Ye
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry; Beijing Normal University; Beijing 100875 China
| | - Xia Wang
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry; Beijing Normal University; Beijing 100875 China
| | - Winnie Deuther-Conrad
- Helmholtz-Zentrum Dresden-Rossendorf, Department of Neuroradiopharmaceuticals; Institute of Radiopharmaceutical Cancer Research; 04318 Leipzig Germany
| | - Jinming Zhang
- Nuclear Medicine Department; Chinese PLA General Hospital; Beijing 100853 China
| | - Jianzhou Li
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry; Beijing Normal University; Beijing 100875 China
| | - Xiaojun Zhang
- Nuclear Medicine Department; Chinese PLA General Hospital; Beijing 100853 China
| | - Liang Wang
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry; Beijing Normal University; Beijing 100875 China
| | - Jörg Steinbach
- Helmholtz-Zentrum Dresden-Rossendorf, Department of Neuroradiopharmaceuticals; Institute of Radiopharmaceutical Cancer Research; 04318 Leipzig Germany
| | - Peter Brust
- Helmholtz-Zentrum Dresden-Rossendorf, Department of Neuroradiopharmaceuticals; Institute of Radiopharmaceutical Cancer Research; 04318 Leipzig Germany
| | - Hongmei Jia
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry; Beijing Normal University; Beijing 100875 China
| |
Collapse
|
23
|
Wang Y, Guo L, Jiang HF, Zheng LT, Zhang A, Zhen XC. Allosteric Modulation of Sigma-1 Receptors Elicits Rapid Antidepressant Activity. CNS Neurosci Ther 2016; 22:368-77. [PMID: 26854125 PMCID: PMC6492821 DOI: 10.1111/cns.12502] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Revised: 11/27/2015] [Accepted: 11/28/2015] [Indexed: 02/06/2023] Open
Abstract
AIMS Sigma-1 receptors are involved in the pathophysiological process of several neuropsychiatric diseases such as epilepsy, depression. Allosteric modulation represents an important mechanism for receptor functional regulation. In this study, we examined antidepressant activity of the latest identified novel and selective allosteric modulator of sigma-1 receptor 3-methyl-phenyl-2, 3, 4, 5-tetrahydro-1H-benzo[d]azepin-7-ol (SOMCL-668). METHODS AND RESULTS A single administration of SOMCL-668 decreased the immobility time in the forced swimming test (FST) and tailing suspended test in mice, which were abolished by pretreatment of sigma-1 receptor antagonist BD1047. In the chronic unpredicted mild stress (CUMS) model, chronic application of SOMCL-668 rapidly ameliorated anhedonia-like behavior (within a week), accompanying with the enhanced expression of brain-derived neurotrophic factor (BDNF) and phosphorylation of glycogen synthase kinase 3β (GSK3β) (Ser-9) in the hippocampus. SOMCL-668 also rapidly promoted the phosphorylation of GSK3β (Ser-9) in an allosteric manner in vitro. In the cultured primary neurons, SOMCL-668 enhanced the sigma-1 receptor agonist-induced neurite outgrowth and the secretion of BDNF. CONCLUSION SOMCL-668, a novel allosteric modulator of sigma-1 receptors, elicits a potent and rapid acting antidepressant effect. The present data provide the first evidence that allosteric modulation of sigma-1 receptors may represent a new approach for antidepressant drug discovery.
Collapse
Affiliation(s)
- Yun Wang
- Jiangsu Key laboratory for Translational Research for Neuropsychiatric Diseases, The Collaborative Innovation Center for Brain Science, College of Pharmaceutical Sciences, Soochow University, Jiangsu, China
- Jiangsu Key Laboratory of New Drug Research and Clinical Pharmacy, Department of Pharmacology, Xuzhou Medical College, Jiangsu, China
| | - Lin Guo
- Jiangsu Key laboratory for Translational Research for Neuropsychiatric Diseases, The Collaborative Innovation Center for Brain Science, College of Pharmaceutical Sciences, Soochow University, Jiangsu, China
| | - Hua-Feng Jiang
- Jiangsu Key laboratory for Translational Research for Neuropsychiatric Diseases, The Collaborative Innovation Center for Brain Science, College of Pharmaceutical Sciences, Soochow University, Jiangsu, China
| | - Long-Tai Zheng
- Jiangsu Key laboratory for Translational Research for Neuropsychiatric Diseases, The Collaborative Innovation Center for Brain Science, College of Pharmaceutical Sciences, Soochow University, Jiangsu, China
| | - Ao Zhang
- Department of Medicinal chemistry, Shanghai Institute of Material Medica, Chinese Academy of Sciences, Shanghai, China
| | - Xue-Chu Zhen
- Jiangsu Key laboratory for Translational Research for Neuropsychiatric Diseases, The Collaborative Innovation Center for Brain Science, College of Pharmaceutical Sciences, Soochow University, Jiangsu, China
| |
Collapse
|
24
|
Srivats S, Balasuriya D, Pasche M, Vistal G, Edwardson JM, Taylor CW, Murrell-Lagnado RD. Sigma1 receptors inhibit store-operated Ca2+ entry by attenuating coupling of STIM1 to Orai1. J Cell Biol 2016; 213:65-79. [PMID: 27069021 PMCID: PMC4828687 DOI: 10.1083/jcb.201506022] [Citation(s) in RCA: 69] [Impact Index Per Article: 8.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2015] [Accepted: 02/24/2016] [Indexed: 11/24/2022] Open
Abstract
Sigma1 receptors (σ1Rs) are expressed widely; they bind diverse ligands, including psychotropic drugs and steroids, regulate many ion channels, and are implicated in cancer and addiction. It is not known how σ1Rs exert such varied effects. We demonstrate that σ1Rs inhibit store-operated Ca(2+)entry (SOCE), a major Ca(2+)influx pathway, and reduce the Ca(2+)content of the intracellular stores. SOCE was inhibited by expression of σ1R or an agonist of σ1R and enhanced by loss of σ1R or an antagonist. Within the endoplasmic reticulum (ER), σ1R associated with STIM1, the ER Ca(2+)sensor that regulates SOCE. This interaction was modulated by σ1R ligands. After depletion of Ca(2+)stores, σ1R accompanied STIM1 to ER-plasma membrane (PM) junctions where STIM1 stimulated opening of the Ca(2+)channel, Orai1. The association of STIM1 with σ1R slowed the recruitment of STIM1 to ER-PM junctions and reduced binding of STIM1 to PM Orai1. We conclude that σ1R attenuates STIM1 coupling to Orai1 and thereby inhibits SOCE.
Collapse
Affiliation(s)
- Shyam Srivats
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, England, UK
| | - Dilshan Balasuriya
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, England, UK
| | - Mathias Pasche
- MRC Laboratory for Molecular Biology, Cambridge CB2 0QH, England, UK
| | - Gerard Vistal
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, England, UK
| | - J Michael Edwardson
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, England, UK
| | - Colin W Taylor
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, England, UK
| | - Ruth D Murrell-Lagnado
- Department of Pharmacology, University of Cambridge, Cambridge CB2 1PD, England, UK Sussex Neuroscience, School of Life Sciences, University of Sussex, Brighton BN1 9QG, England, UK
| |
Collapse
|
25
|
Su TP, Su TC, Nakamura Y, Tsai SY. The Sigma-1 Receptor as a Pluripotent Modulator in Living Systems. Trends Pharmacol Sci 2016; 37:262-278. [PMID: 26869505 PMCID: PMC4811735 DOI: 10.1016/j.tips.2016.01.003] [Citation(s) in RCA: 233] [Impact Index Per Article: 29.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2015] [Revised: 01/03/2016] [Accepted: 01/05/2016] [Indexed: 01/21/2023]
Abstract
The sigma-1 receptor (Sig-1R) is an endoplasmic reticulum (ER) protein that resides specifically in the mitochondria-associated endoplasmic reticulum (ER) membrane (MAM), an interface between ER and mitochondria. In addition to being able to translocate to the plasma membrane (PM) to interact with ion channels and other receptors, Sig-1R also occurs at the nuclear envelope, where it recruits chromatin-remodeling factors to affect the transcription of genes. Sig-1Rs have also been reported to interact with other membranous or soluble proteins at other loci, including the cytosol, and to be involved in several central nervous system (CNS) diseases. Here, we propose that Sig-1R is a pluripotent modulator with resultant multiple functional manifestations in living systems.
Collapse
Affiliation(s)
- Tsung-Ping Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD 21224, USA.
| | - Tzu-Chieh Su
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD 21224, USA
| | - Yoki Nakamura
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD 21224, USA
| | - Shang-Yi Tsai
- Cellular Pathobiology Section, Integrative Neuroscience Research Branch, Intramural Research Program, National Institute on Drug Abuse, National Institutes of Health, Department of Health and Human Services, Baltimore, MD 21224, USA
| |
Collapse
|
26
|
Shinohara K, Kishi T, Hirooka Y, Sunagawa K. Circulating angiotensin II deteriorates left ventricular function with sympathoexcitation via brain angiotensin II receptor. Physiol Rep 2015; 3:3/8/e12514. [PMID: 26290529 PMCID: PMC4562594 DOI: 10.14814/phy2.12514] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Sympathoexcitation contributes to the progression of heart failure. Activation of brain angiotensin II type 1 receptors (AT1R) causes central sympathoexcitation. Thus, we assessed the hypothesis that the increase in circulating angiotensin II comparable to that reported in heart failure model affects cardiac function through the central sympathoexcitation via activating AT1R in the brain. In Sprague-Dawley rats, the subcutaneous infusion of angiotensin II for 14 days increased the circulating angiotensin II level comparable to that reported in heart failure model rats after myocardial infarction. In comparison with the control, angiotensin II infusion increased 24 hours urinary norepinephrine excretion, and systolic blood pressure. Angiotensin II infusion hypertrophied left ventricular (LV) without changing chamber dimensions while increased end-diastolic pressure. The LV pressure–volume relationship indicated that angiotensin II did not impact on the end-systolic elastance, whereas significantly increased end-diastolic elastance. Chronic intracerebroventricular infusion of AT1R blocker, losartan, attenuated these angiotensin II-induced changes. In conclusion, circulating angiotensin II in heart failure is capable of inducing sympathoexcitation via in part AT1R in the brain, subsequently leading to LV diastolic dysfunction.
Collapse
Affiliation(s)
- Keisuke Shinohara
- Departments of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Takuya Kishi
- Department of Advanced Therapeutics for Cardiovascular Diseases, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Yoshitaka Hirooka
- Department of Cardiovascular Regulation and Therapeutics, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| | - Kenji Sunagawa
- Departments of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan
| |
Collapse
|
27
|
Central nervous system circuits modified in heart failure: pathophysiology and therapeutic implications. Heart Fail Rev 2015; 19:759-79. [PMID: 24573960 DOI: 10.1007/s10741-014-9427-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
The pathophysiology of heart failure (HF) is characterized by an abnormal activation of neurohumoral systems, including the sympathetic nervous and the renin-angiotensin-aldosterone systems, which have long-term deleterious effects on the disease progression. Perpetuation of this neurohumoral activation is partially dependent of central nervous system (CNS) pathways, mainly involving the paraventricular nucleus of the hypothalamus and some regions of the brainstem. Modifications in these integrative CNS circuits result in the attenuation of sympathoinhibitory and exacerbation of sympathoexcitatory pathways. In addition to the regulation of sympathetic outflow, these central pathways coordinate a complex network of agents with an established pathophysiological relevance in HF such as angiotensin, aldosterone, and proinflammatory cytokines. Central pathways could be potential targets in HF therapy since the current mainstay of HF pharmacotherapy aims primarily at antagonizing the peripheral mechanisms. Thus, in the present review, we describe the role of CNS pathways in HF pathophysiology and as potential novel therapeutic targets.
Collapse
|
28
|
Xie F, Bergmann R, Kniess T, Deuther-Conrad W, Mamat C, Neuber C, Liu B, Steinbach J, Brust P, Pietzsch J, Jia H. (18)F-Labeled 1,4-Dioxa-8-azaspiro[4.5]decane Derivative: Synthesis and Biological Evaluation of a σ1 Receptor Radioligand with Low Lipophilicity as Potent Tumor Imaging Agent. J Med Chem 2015; 58:5395-407. [PMID: 26090686 DOI: 10.1021/acs.jmedchem.5b00593] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
We report the syntheses and evaluation of series of novel piperidine compounds with low lipophilicity as σ1 receptor ligands. 8-(4-(2-Fluoroethoxy)benzyl)-1,4-dioxa-8-azaspiro[4.5]decane (5a) possessed high affinity (K(i) = 5.4 ± 0.4 nM) for σ1 receptors and selectivity for σ2 receptors (30-fold) and the vesicular acetylcholine transporter (1404-fold). [(18)F]5a was prepared using a one-pot, two-step labeling procedure in an automated synthesis module, with a radiochemical purity of >95%, and a specific activity of 25-45 GBq/μmol. Cellular association, biodistribution, and autoradiography with blocking experiments indicated specific binding of [(18)F]5a to σ1 receptors in vitro and in vivo. Small animal positron emission tomography (PET) imaging using mouse tumor xenograft models demonstrated a high accumulation in human carcinoma and melanoma. Treatment with haloperidol significantly reduced the accumulation of the radiotracer in tumors. These findings suggest that radiotracer with suitable lipophilicity and appropriate affinity for σ1 receptors could be used for tumor imaging.
Collapse
Affiliation(s)
- Fang Xie
- †Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, POB 510119, D-01314 Dresden, Germany.,‡Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Ralf Bergmann
- †Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, POB 510119, D-01314 Dresden, Germany
| | - Torsten Kniess
- †Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, POB 510119, D-01314 Dresden, Germany
| | - Winnie Deuther-Conrad
- †Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, POB 510119, D-01314 Dresden, Germany
| | - Constantin Mamat
- †Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, POB 510119, D-01314 Dresden, Germany.,§Technische Universität Dresden, Department of Chemistry and Food Chemistry, D-01062 Dresden, Germany
| | - Christin Neuber
- †Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, POB 510119, D-01314 Dresden, Germany
| | - Boli Liu
- ‡Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| | - Jörg Steinbach
- †Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, POB 510119, D-01314 Dresden, Germany.,§Technische Universität Dresden, Department of Chemistry and Food Chemistry, D-01062 Dresden, Germany
| | - Peter Brust
- †Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, POB 510119, D-01314 Dresden, Germany
| | - Jens Pietzsch
- †Helmholtz-Zentrum Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, POB 510119, D-01314 Dresden, Germany.,§Technische Universität Dresden, Department of Chemistry and Food Chemistry, D-01062 Dresden, Germany
| | - Hongmei Jia
- ‡Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, China
| |
Collapse
|
29
|
Abstract
This review compares the biological and physiological function of Sigma receptors [σRs] and their potential therapeutic roles. Sigma receptors are widespread in the central nervous system and across multiple peripheral tissues. σRs consist of sigma receptor one (σ1R) and sigma receptor two (σ2R) and are expressed in numerous regions of the brain. The sigma receptor was originally proposed as a subtype of opioid receptors and was suggested to contribute to the delusions and psychoses induced by benzomorphans such as SKF-10047 and pentazocine. Later studies confirmed that σRs are non-opioid receptors (not an µ opioid receptor) and play a more diverse role in intracellular signaling, apoptosis and metabolic regulation. σ1Rs are intracellular receptors acting as chaperone proteins that modulate Ca2+ signaling through the IP3 receptor. They dynamically translocate inside cells, hence are transmembrane proteins. The σ1R receptor, at the mitochondrial-associated endoplasmic reticulum membrane, is responsible for mitochondrial metabolic regulation and promotes mitochondrial energy depletion and apoptosis. Studies have demonstrated that they play a role as a modulator of ion channels (K+ channels; N-methyl-d-aspartate receptors [NMDAR]; inositol 1,3,5 triphosphate receptors) and regulate lipid transport and metabolism, neuritogenesis, cellular differentiation and myelination in the brain. σ1R modulation of Ca2+ release, modulation of cardiac myocyte contractility and may have links to G-proteins. It has been proposed that σ1Rs are intracellular signal transduction amplifiers. This review of the literature examines the mechanism of action of the σRs, their interaction with neurotransmitters, pharmacology, location and adverse effects mediated through them.
Collapse
Affiliation(s)
- Colin G Rousseaux
- a Department of Pathology and Laboratory Medicine , University of Ottawa , Ottawa , ON , Canada and
| | | |
Collapse
|
30
|
Abstract
INTRODUCTION Neuropathic pain is difficult to relieve with standard analgesics and tends to be resistant to opioid therapy. Sigma-1 receptors activated during neuropathic injury may sustain pain. Neuropathic injury activates sigma-1 receptors, which results in activation of various kinases, modulates the activity of multiple ion channels, ligand activated ion channels and voltage-gated ion channels; alters monoamine neurotransmission and dampens opioid receptors G-protein activation. Activation of sigma-1 receptors tonically inhibits opioid receptor G-protein activation and thus dampens analgesic responses. Therefore, sigma-1 receptor antagonists are potential analgesics for neuropathic and adjuvants to opioid therapy. AREAS COVERED This article reviews the importance of sigma-1 receptors as pain generators in multiple animal models in order to illustrate both the importance of these unique receptors in pathologic pain and the potential benefits to sigma-1 receptor antagonists as analgesics. EXPERT OPINION Sigma-1 receptor antagonists have a great potential as analgesics for acute neuropathic injury (herpes zoster, acute postoperative pain and chemotherapy induced neuropathy) and may, as an additional benefit, prevent the development of chronic neuropathic pain. Antagonists are potentially effective as adjuvants to opioid therapy when used early to prevent analgesic tolerance. Drug development is complicated by the complexity of sigma-1 receptor pharmacodynamics and its multiple targets, the lack of a specific sigma-1 receptor antagonist, and potential side effects due to on-target toxicities (cognitive impairment, depression).
Collapse
Affiliation(s)
- Mellar P Davis
- Case Western Reserve University, Taussig Cancer Institute, Cleveland Clinic Lerner School of Medicine, Palliative Medicine and Supportive Oncology Services, Division of Solid Tumor, The Cleveland Clinic , 9500 Euclid Ave, Cleveland, OH 44195 , USA
| |
Collapse
|
31
|
Chen YY, Wang X, Zhang JM, Deuther-Conrad W, Zhang XJ, Huang Y, Li Y, Ye JJ, Cui MC, Steinbach J, Brust P, Liu BL, Jia HM. Synthesis and evaluation of a 18F-labeled spirocyclic piperidine derivative as promising σ1 receptor imaging agent. Bioorg Med Chem 2014; 22:5270-8. [DOI: 10.1016/j.bmc.2014.08.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2014] [Revised: 07/18/2014] [Accepted: 08/05/2014] [Indexed: 12/16/2022]
|
32
|
Ahola AJ, Harjutsalo V, Forsblom C, Groop PH. Renin-angiotensin-aldosterone-blockade is associated with decreased use of antidepressant therapy in patients with type 1 diabetes and diabetic nephropathy. Acta Diabetol 2014; 51:529-33. [PMID: 24436029 DOI: 10.1007/s00592-013-0547-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/19/2013] [Accepted: 12/12/2013] [Indexed: 01/21/2023]
Abstract
Hypertension and depression are frequent comorbidities of diabetes. Studies suggest that antihypertensive medication affecting the renin-angiotensin-aldosterone system (RAAS) might also relieve depression. Whether this is also seen in patients with type 1 diabetes is not known. We therefore studied whether use of RAAS-modifying medication is associated with reduced antidepressant use in type 1 diabetes. In all, 1,705 participants in the FinnDiane Study were included (57 % men, mean age 46 ± 11 years). Data on medications were obtained from the Drug Prescription Register. Based on their albumin excretion rate (AER), the patients were classified as having normal AER, microalbuminuria, or macroalbuminuria. Diabetic nephropathy was defined as macroalbuminuria or end-stage renal disease (dialysis or renal transplant). A total of 8.4 and 10.9 % of patients with and without RAAS-modifying medication, respectively, had antidepressant medication purchases (NS). In logistic regression analysis, after adjusting for potential confounding factors, use of RAAS-modifying medication was not associated with antidepressant purchases. However, when patients with and without diabetic nephropathy were analyzed separately, RAAS-modifying medication was associated with lower frequency of antidepressant purchases among patients with established diabetic nephropathy. In conclusion, use of RAAS-modifying medication may improve mood in patients with type 1 diabetes and established diabetic nephropathy.
Collapse
Affiliation(s)
- Aila J Ahola
- Folkhälsan Research Center, Folkhälsan Institute of Genetics, Biomedicum Helsinki C318b, University of Helsinki, PO Box 63, 00014, Helsinki, Finland
| | | | | | | |
Collapse
|
33
|
Central neuregulin-1/ErbB signaling modulates cardiac function via sympathetic activity in pressure overload-induced heart failure. J Hypertens 2014; 32:817-25. [DOI: 10.1097/hjh.0000000000000072] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
|
34
|
Tagashira H, Bhuiyan MS, Shioda N, Fukunaga K. Fluvoxamine rescues mitochondrial Ca2+ transport and ATP production through σ(1)-receptor in hypertrophic cardiomyocytes. Life Sci 2013; 95:89-100. [PMID: 24373833 DOI: 10.1016/j.lfs.2013.12.019] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/21/2013] [Revised: 11/19/2013] [Accepted: 12/12/2013] [Indexed: 01/15/2023]
Abstract
AIMS We previously reported that fluvoxamine, a selective serotonin reuptake inhibitor with high affinity for the σ1-receptor (σ1R), ameliorates cardiac hypertrophy and dysfunction via σ1R stimulation. Although σ1R on non-cardiomyocytes interacts with the IP3 receptor (IP3R) to promote mitochondrial Ca(2+) transport, little is known about its physiological and pathological relevance in cardiomyocytes. MAIN METHODS Here we performed Ca(2+) imaging and measured ATP production to define the role of σ1Rs in regulating sarcoplasmic reticulum (SR)-mitochondrial Ca(2+) transport in neonatal rat ventricular cardiomyocytes treated with angiotensin II to promote hypertrophy. KEY FINDING These cardiomyocytes exhibited imbalances in expression levels of σ1R and IP3R and impairments in both phenylephrine-induced mitochondrial Ca(2+) mobilization from the SR and ATP production. Interestingly, σ1R stimulation with fluvoxamine rescued impaired mitochondrial Ca(2+) mobilization and ATP production, an effect abolished by treatment of cells with the σ1R antagonist, NE-100. Under physiological conditions, fluvoxamine stimulation of σ1Rs suppressed intracellular Ca(2+) mobilization through IP3Rs and ryanodine receptors (RyRs). In vivo, chronic administration of fluvoxamine to TAC mice also rescued impaired ATP production. SIGNIFICANCE These results suggest that σ1R stimulation with fluvoxamine promotes SR-mitochondrial Ca(2+) transport and mitochondrial ATP production, whereas σ1R stimulation suppresses intracellular Ca(2+) overload through IP3Rs and RyRs. These mechanisms likely underlie in part the anti-hypertrophic and cardioprotective action of the σ1R agonists including fluvoxamine.
Collapse
Affiliation(s)
- Hideaki Tagashira
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Md Shenuarin Bhuiyan
- Division of Molecular Cardiovascular Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | - Norifumi Shioda
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan
| | - Kohji Fukunaga
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, Sendai, Japan.
| |
Collapse
|
35
|
Honda N, Hirooka Y, Ito K, Matsukawa R, Shinohara K, Kishi T, Yasukawa K, Utsumi H, Sunagawa K. Moxonidine-induced central sympathoinhibition improves prognosis in rats with hypertensive heart failure. J Hypertens 2013; 31:2300-8; discussion 2308. [DOI: 10.1097/hjh.0b013e328364a2a1] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
36
|
Ito K, Hirooka Y, Sunagawa K. Brain sigma-1 receptor stimulation improves mental disorder and cardiac function in mice with myocardial infarction. J Cardiovasc Pharmacol 2013; 62:222-8. [PMID: 23615161 DOI: 10.1097/fjc.0b013e3182970b15] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Mental disorder after myocardial infarction (MI) is reported by many epidemiological studies and is associated with a poor prognosis. The reduction of brain sigma-1 receptor (S1R) plays an important role in the pathogenesis of mental disorder, and we recently demonstrated that the reduction of brain S1R causes sympathoexcitation. However, the role of brain S1R in the association between MI and mental disorder, such as depression or cognitive impairment, remains unclear. To investigate this, we performed left coronary artery ligation on mice to produce an MI model (MI-mice). Compared with sham-operated controls (Sham-mice), MI-mice showed augmented sympathetic activity, decreased cardiac function, and lower S1R expression in both the hypothalamus and hippocampus. Furthermore, MI-mice displayed decreased Y-maze spontaneous alternation (a maker of spatial working memory), decreased circadian variation in locomotor activity, and increased immobility time in the tail suspension test (markers of depression-like behavior). Intracerebroventricular infusion of the S1R agonist PRE084 in MI-mice improved both mental disorder and cardiac function with lowered sympathetic activity and the recovery of the S1R expression in both the hypothalamus and hippocampus. These results indicate that brain S1R is decreased in MI-mice and that this plays an important role in the coexistence of increased heart failure via sympathoexcitation and mental disorders, such as depression or cognitive impairment.
Collapse
Affiliation(s)
- Koji Ito
- Department of Cardiovascular Medicine, Kyushu University Graduate School of Medical Sciences, Fukuoka, Japan.
| | | | | |
Collapse
|
37
|
Zheng Z, Zeng Y, Wu J. Increased neuroplasticity may protect against cardiovascular disease. Int J Neurosci 2013; 123:599-608. [PMID: 23510138 DOI: 10.3109/00207454.2013.785949] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Neuroplasticity refers to the capacity of the nervous system to modify its organization such that the brain can be shaped by environmental input. Individuals exhibit different degrees of neuroplasticity because of their different courses of growth. Neuroplasticity may thus play a role in individual differences in the treatment of neuropsychiatric diseases. The nervous system monitors and coordinates internal organ function. Thus neuroplasticity may also be associated with the pathogenesis and the treatment of some other diseases besides neuropsychiatric diseases. The cardiovascular system is controlled by the nervous system, mainly by the autonomic nervous system. Stress may lead to depression and cardiovascular disease (CVD). CVD is associated with depression, which is a disorder of decreased neuroplasticity. And the mechanisms of depression and CVD are related. So we conclude that decreased neuroplasticity causes the coexistence of depression with CVD, and increased neuroplasticity may be beneficial against the development of CVD. This theory provides another angle that can explain some of the reported phenomena related to CVD and neuropsychiatry and provide a potential treatment to protect against CVD.
Collapse
Affiliation(s)
- Zhihua Zheng
- Guangdong Province Pharmaceutical Association, Guangzhou, China.
| | | | | |
Collapse
|
38
|
Li Y, Wang X, Zhang J, Deuther-Conrad W, Xie F, Zhang X, Liu J, Qiao J, Cui M, Steinbach J, Brust P, Liu B, Jia H. Synthesis and Evaluation of Novel 18F-Labeled Spirocyclic Piperidine Derivatives as σ1 Receptor Ligands for Positron Emission Tomography Imaging. J Med Chem 2013; 56:3478-91. [DOI: 10.1021/jm301734g] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Affiliation(s)
- Yan Li
- Key Laboratory
of Radiopharmaceuticals
(Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, People’s
Republic of China
| | - Xia Wang
- Key Laboratory
of Radiopharmaceuticals
(Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, People’s
Republic of China
| | - Jinming Zhang
- Nuclear
Medicine Department, Chinese PLA General Hospital, Beijing 100853, People’s
Republic of China
| | - Winnie Deuther-Conrad
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Research
Site Leipzig, Department of Neuroradiopharmaceuticals, 04318 Leipzig,
Germany
| | - Fang Xie
- Key Laboratory
of Radiopharmaceuticals
(Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, People’s
Republic of China
| | - Xiaojun Zhang
- Nuclear
Medicine Department, Chinese PLA General Hospital, Beijing 100853, People’s
Republic of China
| | - Jian Liu
- Nuclear
Medicine Department, Chinese PLA General Hospital, Beijing 100853, People’s
Republic of China
| | - Jinping Qiao
- Key Laboratory
of Radiopharmaceuticals
(Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, People’s
Republic of China
| | - Mengchao Cui
- Key Laboratory
of Radiopharmaceuticals
(Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, People’s
Republic of China
| | - Jörg Steinbach
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Research
Site Leipzig, Department of Neuroradiopharmaceuticals, 04318 Leipzig,
Germany
| | - Peter Brust
- Helmholtz-Zentrum
Dresden-Rossendorf, Institute of Radiopharmaceutical Cancer Research, Research
Site Leipzig, Department of Neuroradiopharmaceuticals, 04318 Leipzig,
Germany
| | - Boli Liu
- Key Laboratory
of Radiopharmaceuticals
(Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, People’s
Republic of China
| | - Hongmei Jia
- Key Laboratory
of Radiopharmaceuticals
(Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, People’s
Republic of China
| |
Collapse
|
39
|
Depression and cardiac disease: epidemiology, mechanisms, and diagnosis. Cardiovasc Psychiatry Neurol 2013; 2013:695925. [PMID: 23653854 PMCID: PMC3638710 DOI: 10.1155/2013/695925] [Citation(s) in RCA: 217] [Impact Index Per Article: 19.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/29/2013] [Accepted: 03/18/2013] [Indexed: 12/13/2022] Open
Abstract
In patients with cardiovascular disease (CVD), depression is common, persistent, and associated with worse health-related quality of life, recurrent cardiac events, and mortality. Both physiological and behavioral factors—including endothelial dysfunction, platelet abnormalities, inflammation, autonomic nervous system dysfunction, and reduced engagement in health-promoting activities—may link depression with adverse cardiac outcomes. Because of the potential impact of depression on quality of life and cardiac outcomes, the American Heart Association has recommended routine depression screening of all cardiac patients with the 2- and 9-item Patient Health Questionnaires. However, despite the availability of these easy-to-use screening tools and effective treatments, depression is underrecognized and undertreated in patients with CVD. In this paper, we review the literature on epidemiology, phenomenology, comorbid conditions, and risk factors for depression in cardiac disease. We outline the associations between depression and cardiac outcomes, as well as the mechanisms that may mediate these links. Finally, we discuss the evidence for and against routine depression screening in patients with CVD and make specific recommendations for when and how to assess for depression in this high-risk population.
Collapse
|
40
|
Bhuiyan MS, Tagashira H, Fukunaga K. Crucial interactions between selective serotonin uptake inhibitors and sigma-1 receptor in heart failure. J Pharmacol Sci 2013; 121:177-84. [PMID: 23428811 DOI: 10.1254/jphs.12r13cp] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022] Open
Abstract
Depression is associated with a substantial increase in the risk of developing heart failure and is independently associated with increased cardiovascular morbidity and mortality. Inversely, cardiovascular disease can lead to severe depression. Thus, therapy with selective serotonin reuptake inhibitors (SSRIs) is strongly recommended to reduce cardiovascular disease-induced morbidity and mortality. However, molecular mechanisms to support evidence-based SSRI treatment of cardiovascular disease have not been elucidated. We recently found very high expression of the sigma-1 receptor, an orphan receptor, in rat heart tissue and defined the cardiac sigma-1 receptor as a direct SSRI target in eliciting cardioprotection in both pressure overload (PO)induced and transverse aortic constriction (TAC)-induced myocardial hypertrophy models in rodents. Our findings suggest that SSRIs such as fluvoxamine protect against PO- and TAC-induced cardiac dysfunction by upregulating sigma-1 receptor expression and stimulating sigma-1 receptor-mediated Akt-eNOS signaling. Here, we discuss the association of depression and cardiovascular diseases, the protective mechanism of SSRIs in heart failure patients, and the pathophysiological relevance of sigma-1 receptors to progression of heart failure. These findings should promote development of clinical therapeutics targeting the sigma-1 receptor in cardiovascular diseases.
Collapse
Affiliation(s)
- Md Shenuarin Bhuiyan
- Division of Molecular Cardiovascular Biology, Department of Pediatrics, The Cincinnati Children's Hospital Medical Center, Cincinnati, OH 45229, USA
| | | | | |
Collapse
|
41
|
Tagashira H, Zhang C, Lu YM, Hasegawa H, Kanai H, Han F, Fukunaga K. Stimulation of σ1-receptor restores abnormal mitochondrial Ca²⁺ mobilization and ATP production following cardiac hypertrophy. Biochim Biophys Acta Gen Subj 2013; 1830:3082-94. [PMID: 23298811 DOI: 10.1016/j.bbagen.2012.12.029] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/02/2012] [Revised: 12/21/2012] [Accepted: 12/26/2012] [Indexed: 10/27/2022]
Abstract
BACKGROUND We previously reported that the σ1-receptor (σ1R) is down-regulated following cardiac hypertrophy and dysfunction in transverse aortic constriction (TAC) mice. Here we address how σ1R stimulation with the selective σ1R agonist SA4503 restores hypertrophy-induced cardiac dysfunction through σ1R localized in the sarcoplasmic reticulum (SR). METHODS We first confirmed anti-hypertrophic effects of SA4503 (0.1-1μM) in cultured cardiomyocytes exposed to angiotensin II (Ang II). Then, to confirm the ameliorative effects of σ1R stimulation in vivo, we administered SA4503 (1.0mg/kg) and the σ1R antagonist NE-100 (1.0mg/kg) orally to TAC mice for 4weeks (once daily). RESULTS σ1R stimulation with SA4503 significantly inhibited Ang II-induced cardiomyocyte hypertrophy. Ang II exposure for 72h impaired phenylephrine (PE)-induced Ca(2+) mobilization from the SR into both the cytosol and mitochondria. Treatment of cardiomyocytes with SA4503 largely restored PE-induced Ca(2+) mobilization into mitochondria. Exposure of cardiomyocytes to Ang II for 72h decreased basal ATP content and PE-induced ATP production concomitant with reduced mitochondrial size, while SA4503 treatment completely restored ATP production and mitochondrial size. Pretreatment with NE-100 or siRNA abolished these effects. Chronic SA4503 administration also significantly attenuated myocardial hypertrophy and restored ATP production in TAC mice. SA4503 administration also decreased hypertrophy-induced impairments in LV contractile function. CONCLUSIONS σ1R stimulation with the specific agonist SA4503 ameliorates cardiac hypertrophy and dysfunction by restoring both mitochondrial Ca(2+) mobilization and ATP production via σ1R stimulation. GENERAL SIGNIFICANCE Our observations suggest that σ1R stimulation represents a new therapeutic strategy to rescue the heart from hypertrophic dysfunction.
Collapse
Affiliation(s)
- Hideaki Tagashira
- Department of Pharmacology, Graduate School of Pharmaceutical Sciences, Tohoku University, 6-3 Aramaki-Aoba, Aoba-ku, Sendai, Japan
| | | | | | | | | | | | | |
Collapse
|
42
|
Hashimoto K. Sigma-1 receptor chaperone and brain-derived neurotrophic factor: emerging links between cardiovascular disease and depression. Prog Neurobiol 2013; 100:15-29. [PMID: 23044468 DOI: 10.1016/j.pneurobio.2012.09.001] [Citation(s) in RCA: 151] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Revised: 08/31/2012] [Accepted: 09/17/2012] [Indexed: 01/12/2023]
Abstract
Epidemiological studies have demonstrated a close relationship between depression and cardiovascular disease (CVD). Although it is known that the central nervous system (CNS) contributes to this relationship, the detailed mechanisms involved in this process remain unclear. Recent studies suggest that the endoplasmic reticulum (ER) molecular chaperone sigma-1 receptor and brain-derived neurotrophic factor (BDNF) play a role in the pathophysiology of CVD and depression. Several meta-analysis studies have showed that levels of BDNF in the blood of patients with major depressive disorder (MDD) are lower than normal controls, indicating that blood BDNF might be a biomarker for depression. Furthermore, blood levels of BDNF in patients with CVD are also lower than normal controls. A recent study using conditional BDNF knock-out mice in animal models of myocardial infarction highlighted the role of CNS-mediated mechanisms in the cardioprotective effects of BDNF. In addition, a recent study shows that decreased levels of sigma-1 receptor in the mouse brain contribute to the association between heart failure and depression. Moreover, sigma-1 receptor agonists, including the endogenous neurosteroid dehydroepiandosterone (DHEA) and the selective serotonin reuptake inhibitor (SSRI) fluvoxamine, show potent cardioprotective and antidepressive effects in rodents, via sigma-1 receptor stimulation. Interestingly, agonist activation of sigma-1 receptors increased the secretion of mature BDNF from its precursor proBDNF via chaperone activity in the ER. Given the role of ER stress in the pathophysiology of CVD and MDD, the author will discuss the potential link between sigma-1 receptors and BDNF-TrkB pathway in the pathophysiology of these two diseases. Finally, the author will make a case for potent sigma-1 receptor agonists and TrkB agonists as new potential therapeutic drugs for depressive patients with CVD.
Collapse
Affiliation(s)
- Kenji Hashimoto
- Division of Clinical Neuroscience, Chiba University Center for Forensic Mental Health, Inohana, Chiba, Japan.
| |
Collapse
|
43
|
Wang X, Li Y, Deuther-Conrad W, Xie F, Chen X, Cui MC, Zhang XJ, Zhang JM, Steinbach J, Brust P, Liu BL, Jia HM. Synthesis and biological evaluation of ¹⁸F labeled fluoro-oligo-ethoxylated 4-benzylpiperazine derivatives for sigma-1 receptor imaging. Bioorg Med Chem 2013; 21:215-22. [PMID: 23199475 DOI: 10.1016/j.bmc.2012.10.038] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2012] [Revised: 10/11/2012] [Accepted: 10/23/2012] [Indexed: 11/20/2022]
Abstract
We report the synthesis and evaluation of a series of fluoro-oligo-ethoxylated 4-benzylpiperazine derivatives as potential σ(1) receptor ligands. In vitro competition binding assays showed that 1-(1,3-benzodioxol-5-ylmethyl)-4-(4-(2-fluoroethoxy)benzyl)piperazine (6) exhibits low nanomolar affinity for σ(1) receptors (K(i)=1.85 ± 1.59 nM) and high subtype selectivity (σ(2) receptor: K(i)=291 ± 111 nM; K(i)σ(2)/K(i)σ(1)=157). [(18)F]6 was prepared in 30-50% isolated radiochemical yield, with radiochemical purity of >99% by HPLC analysis after purification, via nucleophilic (18)F(-) substitution of the corresponding tosylate precursor. The logD(pH 7.4) value of [(18)F]6 was found to be 2.57 ± 0.10, which is within the range expected to give high brain uptake. Biodistribution studies in mice demonstrated relatively high concentration of radiotracers in organs known to contain σ(1) receptors, including the brain, lungs, kidneys, heart, and spleen. Administration of haloperidol 5 min prior to injection of [(18)F]6 significantly reduced the concentration of radiotracers in the above-mentioned organs. The accumulation of radiotracers in the bone was quite low suggesting that [(18)F]6 is relatively stable to in vivo defluorination. The ex vivo autoradiography in rat brain showed high accumulation of radiotracers in the brain areas known to possess high expression of σ(1) receptors. These findings suggest that [(18)F]6 is a suitable radiotracer for imaging σ(1) receptors with PET in vivo.
Collapse
Affiliation(s)
- Xia Wang
- Key Laboratory of Radiopharmaceuticals (Beijing Normal University), Ministry of Education, College of Chemistry, Beijing Normal University, Beijing 100875, PR China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|