1
|
Pareek A, Singhal R, Pareek A, Ghazi T, Kapoor DU, Ratan Y, Singh AK, Jain V, Chuturgoon AA. Retinoic acid in Parkinson's disease: Molecular insights, therapeutic advances, and future prospects. Life Sci 2024; 355:123010. [PMID: 39181315 DOI: 10.1016/j.lfs.2024.123010] [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: 05/27/2024] [Revised: 08/17/2024] [Accepted: 08/21/2024] [Indexed: 08/27/2024]
Abstract
Parkinson's disease (PD) is a common and progressively worsening neurodegenerative disorder characterized by abnormal protein homeostasis and the degeneration of dopaminergic neurons, particularly in the substantia nigra pars compacta. The prevalence of PD has doubled in the past 25 years, now affecting over 8.5 million individuals worldwide, underscoring the need for effective management strategies. While current pharmacological therapies provide symptom relief, they face challenges in treating advanced PD stages. Recent research highlights the therapeutic benefits of retinoic acid (RA) in PD, demonstrating its potential to mitigate neuroinflammation and oxidative stress, regulate brain aging, promote neuronal plasticity, and influence circadian rhythm gene expression and retinoid X receptor heterodimerization. Additionally, RA helps maintain intestinal homeostasis and modulates the enteric nervous system, presenting significant therapeutic potential for managing PD. This review explores RA as a promising alternative to conventional therapies by summarizing the molecular mechanisms underlying its role in PD pathophysiology and presenting up-to-date insights into both preclinical and clinical studies of RA in PD treatment. It also delves into cutting-edge formulations incorporating RA, highlighting ongoing efforts to refine therapeutic strategies by integrating RA into novel treatments. This comprehensive overview aims to advance progress in the field, contribute to the development of effective, targeted treatments for PD, and enhance patient well-being. Further research is essential to fully explore RA's therapeutic potential and validate its efficacy in PD treatment.
Collapse
Affiliation(s)
- Ashutosh Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, India.
| | - Runjhun Singhal
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, India
| | - Aaushi Pareek
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, India
| | - Terisha Ghazi
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4041, South Africa
| | | | - Yashumati Ratan
- Department of Pharmacy, Banasthali Vidyapith, Banasthali 304022, India
| | - Arun Kumar Singh
- Department of Pharmacy, Vivekananda Global University, Jaipur 303012, India
| | - Vivek Jain
- Department of Pharmaceutical Sciences, Mohanlal Sukhadia University, Udaipur 313001, India
| | - Anil A Chuturgoon
- Discipline of Medical Biochemistry, School of Laboratory Medicine and Medical Sciences, University of KwaZulu-Natal, Durban 4041, South Africa.
| |
Collapse
|
2
|
Yu SJ, Wang Y, Shen H, Bae EK, Li Y, Sambamurti K, Tones MA, Zaleska MM, Hoffer BJ, Greig NH. DPP-4 inhibitors sitagliptin and PF-00734,200 mitigate dopaminergic neurodegeneration, neuroinflammation and behavioral impairment in the rat 6-OHDA model of Parkinson's disease. GeroScience 2024; 46:4349-4371. [PMID: 38563864 PMCID: PMC11336009 DOI: 10.1007/s11357-024-01116-0] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/12/2023] [Accepted: 02/28/2024] [Indexed: 04/04/2024] Open
Abstract
Epidemiological studies report an elevated risk of Parkinson's disease (PD) in patients with type 2 diabetes mellitus (T2DM) that is mitigated in those prescribed dipeptidyl peptidase 4 (DPP-4) inhibitors. With an objective to characterize clinically translatable doses of DPP-4 inhibitors (gliptins) in a well-characterized PD rodent model, sitagliptin, PF-00734,200 or vehicle were orally administered to rats initiated either 7-days before or 7-days after unilateral medial forebrain bundle 6-hydroxydopamine (6-OHDA) lesioning. Measures of dopaminergic cell viability, dopamine content, neuroinflammation and neurogenesis were evaluated thereafter in ipsi- and contralateral brain. Plasma and brain incretin and DPP-4 activity levels were quantified. Furthermore, brain incretin receptor levels were age-dependently evaluated in rodents, in 6-OHDA challenged animals and human subjects with/without PD. Cellular studies evaluated neurotrophic/neuroprotective actions of combined incretin administration. Pre-treatment with oral sitagliptin or PF-00734,200 reduced methamphetamine (meth)-induced rotation post-lesioning and dopaminergic degeneration in lesioned substantia nigra pars compacta (SNc) and striatum. Direct intracerebroventricular gliptin administration lacked neuroprotective actions, indicating that systemic incretin-mediated mechanisms underpin gliptin-induced favorable brain effects. Post-treatment with a threefold higher oral gliptin dose, likewise, mitigated meth-induced rotation, dopaminergic neurodegeneration and neuroinflammation, and augmented neurogenesis. These gliptin-induced actions associated with 70-80% plasma and 20-30% brain DPP-4 inhibition, and elevated plasma and brain incretin levels. Brain incretin receptor protein levels were age-dependently maintained in rodents, preserved in rats challenged with 6-OHDA, and in humans with PD. Combined GLP-1 and GIP receptor activation in neuronal cultures resulted in neurotrophic/neuroprotective actions superior to single agonists alone. In conclusion, these studies support further evaluation of the repurposing of clinically approved gliptins as a treatment strategy for PD.
Collapse
Affiliation(s)
- Seong-Jin Yu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, 35053, Taiwan.
- National Institute On Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA.
| | - Hui Shen
- National Institute On Drug Abuse, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Eun-Kyung Bae
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, 35053, Taiwan
| | - Yazhou Li
- National Institute On Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA
| | - Kumar Sambamurti
- Department of Neurosciences, the Medical University of South Carolina, Charleston, SC, 29425, USA
| | | | | | - Barry J Hoffer
- Department of Neurosurgery, University Hospitals of Cleveland, Case Western Reserve University School of Medicine, Cleveland, OH, 44106, USA
| | - Nigel H Greig
- National Institute On Aging, Intramural Research Program, National Institutes of Health, Baltimore, MD, 21224, USA.
| |
Collapse
|
3
|
Kola A, Nencioni F, Valensin D. Bioinorganic Chemistry of Micronutrients Related to Alzheimer's and Parkinson's Diseases. Molecules 2023; 28:5467. [PMID: 37513339 PMCID: PMC10385134 DOI: 10.3390/molecules28145467] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2023] [Revised: 07/10/2023] [Accepted: 07/13/2023] [Indexed: 07/30/2023] Open
Abstract
Metal ions are fundamental to guarantee the regular physiological activity of the human organism. Similarly, vitamins play a key role in many biological functions of the metabolism, among which are coenzymes, redox mediators, and antioxidants. Due to their importance in the human organism, both metals and vitamins have been extensively studied for their involvement in neurodegenerative diseases (NDs). However, the full potential of the interaction between vitamins and metal ions has not been fully explored by researchers yet, and further investigation on this topic is needed. The aim of this review is to provide an overview of the scientific literature on the implications of vitamins and selected metal ions in two of the most common neurodegenerative diseases, Alzheimer's and Parkinson's disease. Furthermore, vitamin-metal ion interactions are discussed in detail focusing on their bioinorganic chemistry, with the perspective of arousing more interest in this fascinating bioinorganic field.
Collapse
Affiliation(s)
| | | | - Daniela Valensin
- Department of Biotechnology, Chemistry and Pharmacy, University of Siena, Via Aldo Moro 2, 53100 Siena, Italy; (A.K.); (F.N.)
| |
Collapse
|
4
|
Could Vitamins Have a Positive Impact on the Treatment of Parkinson's Disease? Brain Sci 2023; 13:brainsci13020272. [PMID: 36831815 PMCID: PMC9954544 DOI: 10.3390/brainsci13020272] [Citation(s) in RCA: 4] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/31/2022] [Revised: 01/28/2023] [Accepted: 02/02/2023] [Indexed: 02/10/2023] Open
Abstract
Parkinson's disease (PD) is the second most common progressive neurodegenerative disorder after Alzheimer's disease. Pathophysiologically, it is characterized by intracytoplasmic aggregates of α-synuclein protein in the Lewy body and loss of dopaminergic neurons from substantia nigra pars compacta and striatum regions of the brain. Although the exact mechanism of neurodegeneration is not fully elucidated, it has been reported that environmental toxins such as MPTP, rotenone, paraquat, and MPP+ induce oxidative stress, which is one of the causative factors for it. To date, there is no complete cure. However, the indispensable role of oxidative stress in mediating PD indicates that antioxidant therapy could be a possible therapeutic strategy against the disease. The deficiency of vitamins has been extensively co-related to PD. Dietary supplementation of vitamins with antioxidant, anti-inflammatory, anti-apoptotic, and free radical scavenging properties could be the potential neuroprotective therapeutic strategy. This review summarizes the studies that evaluated the role of vitamins (A, B, C, D, E, and K) in PD. It will guide future studies in understanding the potential therapeutic role of vitamins in disease pathophysiology and may provide a framework for designing treatment strategies against the disease.
Collapse
|
5
|
Paccione N, Rahmani M, Barcia E, Negro S. Antiparkinsonian Agents in Investigational Polymeric Micro- and Nano-Systems. Pharmaceutics 2022; 15:pharmaceutics15010013. [PMID: 36678642 PMCID: PMC9866990 DOI: 10.3390/pharmaceutics15010013] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2022] [Revised: 12/13/2022] [Accepted: 12/15/2022] [Indexed: 12/24/2022] Open
Abstract
Parkinson's disease (PD) is a devastating neurodegenerative disease characterized by progressive destruction of dopaminergic tissue in the central nervous system (CNS). To date, there is no cure for the disease, with current pharmacological treatments aimed at controlling the symptoms. Therefore, there is an unmet need for new treatments for PD. In addition to new therapeutic options, there exists the need for improved efficiency of the existing ones, as many agents have difficulties in crossing the blood-brain barrier (BBB) to achieve therapeutic levels in the CNS or exhibit inappropriate pharmacokinetic profiles, thereby limiting their clinical benefits. To overcome these limitations, an interesting approach is the use of drug delivery systems, such as polymeric microparticles (MPs) and nanoparticles (NPs) that allow for the controlled release of the active ingredients targeting to the desired site of action, increasing the bioavailability and efficacy of treatments, as well as reducing the number of administrations and adverse effects. Here we review the polymeric micro- and nano-systems under investigation as potential new therapies for PD.
Collapse
Affiliation(s)
- Nicola Paccione
- Department of Pharmaceutics and Food Technology, School of Pharmacy, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Mahdieh Rahmani
- Department of Pharmaceutics and Food Technology, School of Pharmacy, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
- Correspondence: ; Tel.: +34-913941741
| | - Emilia Barcia
- Department of Pharmaceutics and Food Technology, School of Pharmacy, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
- Institute of Industrial Pharmacy, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| | - Sofía Negro
- Department of Pharmaceutics and Food Technology, School of Pharmacy, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
- Institute of Industrial Pharmacy, Universidad Complutense de Madrid, Ciudad Universitaria s/n, 28040 Madrid, Spain
| |
Collapse
|
6
|
Marie A, Leroy J, Darricau M, Alfos S, De Smedt-Peyrusse V, Richard E, Vancassel S, Bosch-Bouju C. Preventive Vitamin A Supplementation Improves Striatal Function in 6-Hydroxydopamine Hemiparkinsonian Rats. Front Nutr 2022; 9:811843. [PMID: 35178422 PMCID: PMC8843942 DOI: 10.3389/fnut.2022.811843] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2021] [Accepted: 01/05/2022] [Indexed: 12/02/2022] Open
Abstract
Background The mechanisms leading to a loss of dopaminergic (DA) neurons from the substantia nigra pars compacta (SNc) in Parkinson's disease (PD) have multifactorial origins. In this context, nutrition is currently investigated as a modifiable environmental factor for the prevention of PD. In particular, initial studies revealed the deleterious consequences of vitamin A signaling failure on dopamine-related motor behaviors. However, the potential of vitamin A supplementation itself to prevent neurodegeneration has not been established yet. Objective The hypothesis tested in this study is that preventive vitamin A supplementation can protect DA neurons in a rat model of PD. Methods The impact of a 5-week preventive supplementation with vitamin A (20 IU/g of diet) was measured on motor and neurobiological alterations induced by 6-hydroxydopamine (6-OHDA) unilateral injections in the striatum of rats. Rotarod, step test and cylinder tests were performed up to 3 weeks after the lesion. Post-mortem analyses (retinol and monoamines dosages, western blots, immunofluorescence) were performed to investigate neurobiological processes. Results Vitamin A supplementation improved voluntary movements in the cylinder test. In 6-OHDA lesioned rats, a marked decrease of dopamine levels in striatum homogenates was measured. Tyrosine hydroxylase labeling in the SNc and in the striatum was significantly decreased by 6-OHDA injection, without effect of vitamin A. By contrast, vitamin A supplementation increased striatal expression of D2 and RXR receptors in the striatum of 6-OHDA lesioned rats. Conclusions Vitamin A supplementation partially alleviates motor alterations and improved striatal function, revealing a possible beneficial preventive approach for PD.
Collapse
Affiliation(s)
- Anaïs Marie
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Institut Polytechnique de Bordeaux (INP), NutriNeuro, University of Bordeaux, Bordeaux, France
| | - Julien Leroy
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Institut Polytechnique de Bordeaux (INP), NutriNeuro, University of Bordeaux, Bordeaux, France
| | - Morgane Darricau
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Institut Polytechnique de Bordeaux (INP), NutriNeuro, University of Bordeaux, Bordeaux, France
- Institut des Maladies Neurodégénératives, University of Bordeaux, Bordeaux, France
| | - Serge Alfos
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Institut Polytechnique de Bordeaux (INP), NutriNeuro, University of Bordeaux, Bordeaux, France
| | - Veronique De Smedt-Peyrusse
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Institut Polytechnique de Bordeaux (INP), NutriNeuro, University of Bordeaux, Bordeaux, France
| | - Emmanuel Richard
- Institut National de la Santé et de la Recherche Médicale (INSERM), Centre Hospital-Universitaire (CHU) Bordeaux, University of Bordeaux, Bordeaux, France
| | - Sylvie Vancassel
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Institut Polytechnique de Bordeaux (INP), NutriNeuro, University of Bordeaux, Bordeaux, France
| | - Clementine Bosch-Bouju
- Institut National de Recherche pour l'Agriculture, l'Alimentation et l'Environnement (INRAE), Institut Polytechnique de Bordeaux (INP), NutriNeuro, University of Bordeaux, Bordeaux, France
- *Correspondence: Clementine Bosch-Bouju
| |
Collapse
|
7
|
Rai SN, Singh P, Steinbusch HW, Vamanu E, Ashraf G, Singh MP. The Role of Vitamins in Neurodegenerative Disease: An Update. Biomedicines 2021; 9:1284. [PMID: 34680401 PMCID: PMC8533313 DOI: 10.3390/biomedicines9101284] [Citation(s) in RCA: 49] [Impact Index Per Article: 16.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2021] [Revised: 09/14/2021] [Accepted: 09/15/2021] [Indexed: 02/06/2023] Open
Abstract
Acquiring the recommended daily allowance of vitamins is crucial for maintaining homeostatic balance in humans and other animals. A deficiency in or dysregulation of vitamins adversely affects the neuronal metabolism, which may lead to neurodegenerative diseases. In this article, we discuss how novel vitamin-based approaches aid in attenuating abnormal neuronal functioning in neurodegeneration-based brain diseases such as Alzheimer's disease, Parkinson's disease, Huntington's disease, Amyotrophic lateral sclerosis, and Prion disease. Vitamins show their therapeutic activity in Parkinson's disease by antioxidative and anti-inflammatory activity. In addition, different water- and lipid-soluble vitamins have also prevented amyloid beta and tau pathology. On the other hand, some results also show no correlation between vitamin action and the prevention of neurodegenerative diseases. Some vitamins also exhibit toxic activity too. This review discusses both the beneficial and null effects of vitamin supplementation for neurological disorders. The detailed mechanism of action of both water- and lipid-soluble vitamins is addressed in the manuscript. Hormesis is also an essential factor that is very helpful to determine the effective dose of vitamins. PubMed, Google Scholar, Web of Science, and Scopus were employed to conduct the literature search of original articles, review articles, and meta-analyses.
Collapse
Affiliation(s)
- Sachchida Nand Rai
- Centre of Biotechnology, University of Allahabad, Prayagraj 211002, India;
| | - Payal Singh
- Department of Zoology, MMV, Banaras Hindu University, Varanasi 221005, India;
| | - Harry W.M. Steinbusch
- Department of Cellular Neuroscience, Faculty of Health, Medicine & Life Sciences, Maastricht University, 6211 LK Maastricht, The Netherlands;
- Department of Cognitive Neuroscience, DGIST, Daegu 42988, Korea
| | - Emanuel Vamanu
- Faculty of Biotechnology, The University of Agronomic Science and Veterinary Medicine, 59 Marasti blvd, 1 District, 011464 Bucharest, Romania
| | - Ghulam Ashraf
- Pre-Clinical Research Unit, King Fahd Medical Research Center, King Abdulaziz University, Jeddah 21589, Saudi Arabia;
- Department of Medical Laboratory Technology, Faculty of Applied Medical Sciences, King Abdulaziz University, Jeddah 21589, Saudi Arabia
| | - Mohan Prasad Singh
- Centre of Biotechnology, University of Allahabad, Prayagraj 211002, India;
| |
Collapse
|
8
|
Shin EJ, Jeong JH, Hwang Y, Sharma N, Dang DK, Nguyen BT, Nah SY, Jang CG, Bing G, Nabeshima T, Kim HC. Methamphetamine-induced dopaminergic neurotoxicity as a model of Parkinson's disease. Arch Pharm Res 2021; 44:668-688. [PMID: 34286473 DOI: 10.1007/s12272-021-01341-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2021] [Accepted: 07/06/2021] [Indexed: 12/01/2022]
Abstract
Parkinson's disease (PD) is a progressive neurodegenerative disease with a high prevalence, approximately 1 % in the elderly population. Numerous studies have demonstrated that methamphetamine (MA) intoxication caused the neurological deficits and nigrostriatal damage seen in Parkinsonian conditions, and subsequent rodent studies have found that neurotoxic binge administration of MA reproduced PD-like features, in terms of its symptomatology and pathology. Several anti-Parkinsonian medications have been shown to attenuate the motor impairments and dopaminergic damage induced by MA. In addition, it has been recognized that mitochondrial dysfunction, oxidative stress, pro-apoptosis, proteasomal/autophagic impairment, and neuroinflammation play important roles in inducing MA neurotoxicity. Importantly, MA neurotoxicity has been shown to share a common mechanism of dopaminergic toxicity with that of PD pathogenesis. This review describes the major findings on the neuropathological features and underlying neurotoxic mechanisms induced by MA and compares them with Parkinsonian pathogenesis. Taken together, it is suggested that neurotoxic binge-type administration of MA in rodents is a valid animal model for PD that may provide knowledge on the neuropathogenesis of PD.
Collapse
Affiliation(s)
- Eun-Joo Shin
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Ji Hoon Jeong
- Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, 06974, Seoul, Republic of Korea
| | - Yeonggwang Hwang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Naveen Sharma
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.,Department of Global Innovative Drugs, Graduate School of Chung-Ang University, College of Medicine, Chung-Ang University, 06974, Seoul, Republic of Korea
| | - Duy-Khanh Dang
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.,Pharmacy Faculty, Can Tho University of Medicine and Pharmacy, 900000, Can Tho City, Vietnam
| | - Bao-Trong Nguyen
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea
| | - Seung-Yeol Nah
- Ginsentology Research Laboratory, Department of Physiology, College of Veterinary Medicine and Bio/Molecular Informatics Center, Konkuk University, 05029, Seoul, Republic of Korea
| | - Choon-Gon Jang
- Department of Pharmacology, School of Pharmacy, Sungkyunkwan University, 16419, Suwon, Republic of Korea
| | - Guoying Bing
- Department of Neuroscience, College of Medicine, University of Kentucky, KY, 40536, Lexington, USA
| | - Toshitaka Nabeshima
- Advanced Diagnostic System Research Laboratory, Graduate School of Health Science, Fujita Health University, 470-1192, Toyoake, Japan
| | - Hyoung-Chun Kim
- Neuropsychopharmacology and Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea. .,Neuropsychopharmacology & Toxicology Program, College of Pharmacy, Kangwon National University, 24341, Chunchon, Republic of Korea.
| |
Collapse
|
9
|
Marie A, Darricau M, Touyarot K, Parr-Brownlie LC, Bosch-Bouju C. Role and Mechanism of Vitamin A Metabolism in the Pathophysiology of Parkinson's Disease. JOURNAL OF PARKINSON'S DISEASE 2021; 11:949-970. [PMID: 34120916 PMCID: PMC8461657 DOI: 10.3233/jpd-212671] [Citation(s) in RCA: 18] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Accepted: 05/12/2021] [Indexed: 01/09/2023]
Abstract
Evidence shows that altered retinoic acid signaling may contribute to the pathogenesis and pathophysiology of Parkinson's disease (PD). Retinoic acid is the bioactive derivative of the lipophilic vitamin A. Vitamin A is involved in several important homeostatic processes, such as cell differentiation, antioxidant activity, inflammation and neuronal plasticity. The role of vitamin A and its derivatives in the pathogenesis and pathophysiology of neurodegenerative diseases, and their potential as therapeutics, has drawn attention for more than 10 years. However, the literature sits in disparate fields. Vitamin A could act at the crossroad of multiple environmental and genetic factors of PD. The purpose of this review is to outline what is known about the role of vitamin A metabolism in the pathogenesis and pathophysiology of PD. We examine key biological systems and mechanisms that are under the control of vitamin A and its derivatives, which are (or could be) exploited for therapeutic potential in PD: the survival of dopaminergic neurons, oxidative stress, neuroinflammation, circadian rhythms, homeostasis of the enteric nervous system, and hormonal systems. We focus on the pivotal role of ALDH1A1, an enzyme expressed by dopaminergic neurons for the detoxification of these neurons, which is under the control of retinoic acid. By providing an integrated summary, this review will guide future studies on the potential role of vitamin A in the management of symptoms, health and wellbeing for PD patients.
Collapse
Affiliation(s)
- Anaıs Marie
- University Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Morgane Darricau
- University Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
- University Bordeaux, Institut des Maladies Neurodégénératives, UMR 5293, Bordeaux, France
| | - Katia Touyarot
- University Bordeaux, INRAE, Bordeaux INP, NutriNeuro, UMR 1286, Bordeaux, France
| | - Louise C. Parr-Brownlie
- Department of Anatomy, Brain Health Research Centre, University of Otago, Dunedin, New Zealand
- Brain Research New Zealand (Center of Research Excellence), Dunedin, New Zealand
| | | |
Collapse
|
10
|
Rhea EM, Logsdon AF, Banks WA, Erickson ME. Intranasal Delivery: Effects on the Neuroimmune Axes and Treatment of Neuroinflammation. Pharmaceutics 2020; 12:pharmaceutics12111120. [PMID: 33233734 PMCID: PMC7699866 DOI: 10.3390/pharmaceutics12111120] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2020] [Revised: 11/09/2020] [Accepted: 11/10/2020] [Indexed: 02/02/2023] Open
Abstract
This review highlights the pre-clinical and clinical work performed to use intranasal delivery of various compounds from growth factors to stem cells to reduce neuroimmune interactions. We introduce the concept of intranasal (IN) delivery and the variations of this delivery method based on the model used (i.e., rodents, non-human primates, and humans). We summarize the literature available on IN delivery of growth factors, vitamins and metabolites, cytokines, immunosuppressants, exosomes, and lastly stem cells. We focus on the improvement of neuroimmune interactions, such as the activation of resident central nervous system (CNS) immune cells, expression or release of cytokines, and detrimental effects of signaling processes. We highlight common diseases that are linked to dysregulations in neuroimmune interactions, such as Alzheimer's disease, Parkinson's disease, stroke, multiple sclerosis, and traumatic brain injury.
Collapse
Affiliation(s)
- Elizabeth M. Rhea
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; (A.F.L.); (W.A.B.); (M.E.E.)
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
- Correspondence: ; Tel.: +1-206-764-2938
| | - Aric F. Logsdon
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; (A.F.L.); (W.A.B.); (M.E.E.)
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - William A. Banks
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; (A.F.L.); (W.A.B.); (M.E.E.)
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| | - Michelle E. Erickson
- Geriatrics Research Education and Clinical Center, Veterans Affairs Puget Sound Health Care System, Seattle, WA 98108, USA; (A.F.L.); (W.A.B.); (M.E.E.)
- Division of Gerontology and Geriatric Medicine, Department of Medicine, University of Washington School of Medicine, Seattle, WA 98195, USA
| |
Collapse
|
11
|
Clark JN, Whiting A, McCaffery P. Retinoic acid receptor-targeted drugs in neurodegenerative disease. Expert Opin Drug Metab Toxicol 2020; 16:1097-1108. [DOI: 10.1080/17425255.2020.1811232] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Affiliation(s)
- Jason Nicol Clark
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| | | | - Peter McCaffery
- School of Medicine, Medical Sciences and Nutrition, Institute of Medical Sciences, University of Aberdeen, Aberdeen, Scotland, UK
| |
Collapse
|
12
|
Neuroprotection of GLP-1/GIP receptor agonist via inhibition of mitochondrial stress by AKT/JNK pathway in a Parkinson's disease model. Life Sci 2020; 256:117824. [PMID: 32445758 DOI: 10.1016/j.lfs.2020.117824] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2019] [Revised: 05/10/2020] [Accepted: 05/18/2020] [Indexed: 12/21/2022]
Abstract
OBJECTIVES To investigate the effect of glucagon-like peptide-1 (GLP-1) receptor and glucose dependent insulinotrophic polypeptide (GIP) receptor dual agonist DA-JC4 on alleviating Parkinson's disease (PD) and unveil related cellular mechanisms. METHODS Rotenone was injected to generate a rat PD model, on which the effect of DA-JC4 on motor functions was evaluated by rotational behavioral assay and open field test. The survival of dopaminergic neurons was analyzed, in addition to assays for mitochondrial stress and quantification of neurotransmitter levels using high performance liquid chromatography (HPLC). In cultured hippocampal neurons, the effect of DA-JC4 on mitochondrial stress and related cellular mechanism was analyzed by Flow cytometry, western blotting and reactive oxygen species (ROS). RESULTS DA-JC4 significantly improved motor functions in PD rats, and elevated levels of major neurotransmitters. By histological analysis, DA-JC4 protected dopaminergic neurons from rotenone-induced cell death, which was associated with reduced mitochondrial stress. Experiments in cultured rat hippocampal neurons validated the neuroprotective role of DA-JC4 against cell apoptosis and mitochondrial stress induced by rotenone. The protective effect of DA-JC4 was later found to be dependent on AKT/JNK signal pathway, as treatment using AKT inhibitor or JNK activator abolished such effects. CONCLUSION Our results showed that the dual agonist of GLP-1/GIP receptor could ameliorate motor dysfunctions of PD by protecting dopaminergic neurons which was mediated by relieved mitochondrial stress and apoptosis via AKT/JNK signal pathway.
Collapse
|
13
|
Nuñez-Borque E, González-Naranjo P, Bartolomé F, Alquézar C, Reinares-Sebastián A, Pérez C, Ceballos ML, Páez JA, Campillo NE, Martín-Requero Á. Targeting Cannabinoid Receptor Activation and BACE-1 Activity Counteracts TgAPP Mice Memory Impairment and Alzheimer's Disease Lymphoblast Alterations. Mol Neurobiol 2020; 57:1938-1951. [PMID: 31898159 DOI: 10.1007/s12035-019-01813-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2019] [Accepted: 10/02/2019] [Indexed: 12/20/2022]
Abstract
Alzheimer's disease (AD), the leading cause of dementia in the elderly, is a neurodegenerative disorder marked by progressive impairment of cognitive ability. Patients with AD display neuropathological lesions including senile plaques, neurofibrillary tangles, and neuronal loss. There are no disease-modifying drugs currently available. With the number of affected individuals increasing dramatically throughout the world, there is obvious urgent need for effective treatment strategy for AD. The multifactorial nature of AD encouraged the development of multifunctional compounds, able to interact with several putative targets. Here, we have evaluated the effects of two in-house designed cannabinoid receptors (CB) agonists showing inhibitory actions on β-secretase-1 (BACE-1) (NP137) and BACE-1/butyrylcholinesterase (BuChE) (NP148), on cellular models of AD, including immortalized lymphocytes from late-onset AD patients. Furthermore, the performance of TgAPP mice in a spatial navigation task was investigated following chronic administration of NP137 and NP148. We report here that NP137 and NP148 showed neuroprotective effects in amyloid-β-treated primary cortical neurons, and NP137 in particular rescued the cognitive deficit of TgAPP mice. The latter compound was able to blunt the abnormal cell response to serum addition or withdrawal of lymphoblasts derived from AD patients. It is suggested that NP137 could be a good drug candidate for future treatment of AD.
Collapse
Affiliation(s)
- Emilio Nuñez-Borque
- Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain
| | | | - Fernando Bartolomé
- Neurodegenerative Disorders Group, Instituto de Investigación Hospital 12 de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Carolina Alquézar
- Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain.,Department of Neurology, Memory and Aging Center, University of California, Box 1207, San Francisco, CA, 94158, USA
| | | | | | - Maria L Ceballos
- Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.,Insituto Cajal (CSIC), Madrid, Spain
| | - Juan A Páez
- Instituto de Química Médica (CSIC), Madrid, Spain
| | - Nuria E Campillo
- Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain.
| | - Ángeles Martín-Requero
- Centro de Investigaciones Biológicas (CSIC), Ramiro de Maeztu 9, 28040, Madrid, Spain. .,Centro de Investigación Biomédica en Red sobre Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain.
| |
Collapse
|
14
|
The effects of retinol oral supplementation in 6-hydroxydopamine dopaminergic denervation model in Wistar rats. Neurochem Int 2019; 125:25-34. [DOI: 10.1016/j.neuint.2019.02.002] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2018] [Revised: 02/02/2019] [Accepted: 02/04/2019] [Indexed: 12/23/2022]
|
15
|
Auricular Vagus Nerve Stimulation Exerts Antiinflammatory Effects and Immune Regulatory Function in a 6-OHDA Model of Parkinson’s Disease. Neurochem Res 2018; 43:2155-2164. [DOI: 10.1007/s11064-018-2639-z] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/02/2018] [Revised: 08/03/2018] [Accepted: 09/16/2018] [Indexed: 01/26/2023]
|
16
|
Moutinho M, Codocedo JF, Puntambekar SS, Landreth GE. Nuclear Receptors as Therapeutic Targets for Neurodegenerative Diseases: Lost in Translation. Annu Rev Pharmacol Toxicol 2018; 59:237-261. [PMID: 30208281 DOI: 10.1146/annurev-pharmtox-010818-021807] [Citation(s) in RCA: 29] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Neurodegenerative diseases are characterized by a progressive loss of neurons that leads to a broad range of disabilities, including severe cognitive decline and motor impairment, for which there are no effective therapies. Several lines of evidence support a putative therapeutic role of nuclear receptors (NRs) in these types of disorders. NRs are ligand-activated transcription factors that regulate the expression of a wide range of genes linked to metabolism and inflammation. Although the activation of NRs in animal models of neurodegenerative disease exhibits promising results, the translation of this strategy to clinical practice has been unsuccessful. In this review we discuss the role of NRs in neurodegenerative diseases in light of preclinical and clinical studies, as well as new findings derived from the analysis of transcriptomic databases from humans and animal models. We discuss the failure in the translation of NR-based therapeutic approaches and consider alternative and novel research avenues in the development of effective therapies for neurodegenerative diseases.
Collapse
Affiliation(s)
- Miguel Moutinho
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA;
| | - Juan F Codocedo
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA;
| | - Shweta S Puntambekar
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA;
| | - Gary E Landreth
- Stark Neuroscience Research Institute, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA;
| |
Collapse
|
17
|
Post-treatment with PT302, a long-acting Exendin-4 sustained release formulation, reduces dopaminergic neurodegeneration in a 6-Hydroxydopamine rat model of Parkinson's disease. Sci Rep 2018; 8:10722. [PMID: 30013201 PMCID: PMC6048117 DOI: 10.1038/s41598-018-28449-z] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/06/2018] [Accepted: 06/13/2018] [Indexed: 02/07/2023] Open
Abstract
We previously demonstrated that pretreatment with Exendin-4, a glucagon-like peptide-1 (GLP-1) receptor agonist, reduces 1-methyl-4-phenyl-1,2,3,6-tetrahydropyridine (MPTP) -mediated dopaminergic neurodegeneration. The use of GLP-1 or Exendin-4 for Parkinson's disease (PD) patients is limited by their short half-lives. The purpose of this study was to evaluate a new extended release Exendin-4 formulation, PT302, in a rat model of PD. Subcutaneous administration of PT302 resulted in sustained elevations of Exendin-4 in plasma for >20 days in adult rats. To define an efficacious dose within this range, rats were administered PT302 once every 2 weeks either before or following the unilaterally 6-hydroxydopamine lesioning. Pre- and post-treatment with PT302 significantly reduced methamphetamine-induced rotation after lesioning. For animals given PT302 post lesion, blood and brain samples were collected on day 47 for measurements of plasma Exendin-4 levels and brain tyrosine hydroxylase immunoreactivity (TH-IR). PT302 significantly increased TH-IR in the lesioned substantia nigra and striatum. There was a significant correlation between plasma Exendin-4 levels and TH-IR in the substantia nigra and striatum on the lesioned side. Our data suggest that post-treatment with PT302 provides long-lasting Exendin-4 release and reduces neurodegeneration of nigrostriatal dopaminergic neurons in a 6-hydroxydopamine rat model of PD at a clinically relevant dose.
Collapse
|
18
|
Yang X, Wang Y, Li Q, Zhong Y, Chen L, Du Y, He J, Liao L, Xiong K, Yi CX, Yan J. The Main Molecular Mechanisms Underlying Methamphetamine- Induced Neurotoxicity and Implications for Pharmacological Treatment. Front Mol Neurosci 2018; 11:186. [PMID: 29915529 PMCID: PMC5994595 DOI: 10.3389/fnmol.2018.00186] [Citation(s) in RCA: 132] [Impact Index Per Article: 22.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2018] [Accepted: 05/14/2018] [Indexed: 01/07/2023] Open
Abstract
Methamphetamine (METH) is a popular new-type psychostimulant drug with complicated neurotoxicity. In spite of mounting evidence on METH-induced damage of neural cell, the accurate mechanism of toxic effect of the drug on central nervous system (CNS) has not yet been completely deciphered. Besides, effective treatment strategies toward METH neurotoxicity remain scarce and more efficacious drugs are to be developed. In this review, we summarize cellular and molecular bases that might contribute to METH-elicited neurotoxicity, which mainly include oxidative stress, excitotoxicity, and neuroinflammation. We also discuss some drugs that protect neural cells suffering from METH-induced neurotoxic consequences. We hope more in-depth investigations of exact details that how METH produces toxicity in CNS could be carried out in future and the development of new drugs as natural compounds and immunotherapies, including clinic trials, are expected.
Collapse
Affiliation(s)
- Xue Yang
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yong Wang
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Qiyan Li
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yaxian Zhong
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Liangpei Chen
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Yajun Du
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Jing He
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| | - Lvshuang Liao
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Kun Xiong
- Department of Anatomy and Neurobiology, School of Basic Medical Science, Central South University, Changsha, China
| | - Chun-Xia Yi
- Department of Endocrinology and Metabolism, Academic Medical Center, University of Amsterdam, Amsterdam, Netherlands
| | - Jie Yan
- Department of Forensic Science, School of Basic Medical Sciences, Central South University, Changsha, China
| |
Collapse
|
19
|
Activation of the Cannabinoid Type 2 Receptor by a Novel Indazole Derivative Normalizes the Survival Pattern of Lymphoblasts from Patients with Late-Onset Alzheimer's Disease. CNS Drugs 2018; 32:579-591. [PMID: 29736745 DOI: 10.1007/s40263-018-0515-7] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
BACKGROUND Alzheimer's disease is a multifactorial disorder for which there is no disease-modifying treatment yet. CB2 receptors have emerged as a promising therapeutic target for Alzheimer's disease because they are expressed in neuronal and glial cells and their activation has no psychoactive effects. OBJECTIVE The aim of this study was to investigate whether activation of the CB2 receptor would restore the aberrant enhanced proliferative activity characteristic of immortalized lymphocytes from patients with late-onset Alzheimer's disease. It is assumed that cell-cycle dysfunction occurs in both peripheral cells and neurons in patients with Alzheimer's disease, contributing to the instigation of the disease. METHODS Lymphoblastoid cell lines from patients with Alzheimer's disease and age-matched control individuals were treated with a new, in-house-designed dual drug PGN33, which behaves as a CB2 agonist and butyrylcholinesterase inhibitor. We analyzed the effects of this compound on the rate of cell proliferation and levels of key regulatory proteins. In addition, we investigated the potential neuroprotective action of PGN33 in β-amyloid-treated neuronal cells. RESULTS We report here that PGN33 normalized the increased proliferative activity of Alzheimer's disease lymphoblasts. The compound blunted the calmodulin-dependent overactivation of the PI3K/Akt pathway, by restoring the cyclin-dependent kinase inhibitor p27 levels, which in turn reduced the activity of the cyclin-dependent kinase/pRb cascade. Moreover, this CB2 agonist prevented β-amyloid-induced cell death in neuronal cells. CONCLUSION Our results suggest that the activation of CB2 receptors could be considered a useful therapeutic approach for Alzheimer's disease.
Collapse
|
20
|
Lee PC, Ahmed I, Loriot MA, Mulot C, Paul KC, Bronstein JM, Ritz B, Elbaz A. Smoking and Parkinson disease: Evidence for gene-by-smoking interactions. Neurology 2018; 90:e583-e592. [PMID: 29352099 DOI: 10.1212/wnl.0000000000004953] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2017] [Accepted: 10/20/2017] [Indexed: 12/11/2022] Open
Abstract
OBJECTIVE To investigate whether cigarette smoking interacts with genes involved in individual susceptibility to xenobiotics for the risk of Parkinson disease (PD). METHODS Two French population-based case-control studies (513 patients, 1,147 controls) were included as a discovery sample to examine gene-smoking interactions based on 3,179 single nucleotide polymorphisms (SNPs) in 289 genes involved in individual susceptibility to xenobiotics. SNP-by-cigarette smoking interactions were tested in the discovery sample through an empirical Bayes (EB) approach. Nine SNPs were selected for replication in a population-based case-control study from California (410 patients, 845 controls) with standard logistic regression and the EB approach. For SNPs that replicated, we performed pooled analyses including the discovery and replication datasets and computed pooled odds ratios and confidence intervals (CIs) using random-effects meta-analysis. RESULTS Nine SNPs interacted with smoking in the discovery dataset and were selected for replication. Interactions of smoking with rs4240705 in the RXRA gene and rs1900586 in the SLC17A6 gene were replicated. In pooled analyses (logistic regression), the interactions between smoking and rs4240705-G and rs1900586-G were 1.66 (95% CI 1.28-2.14, p = 1.1 × 10-4, p for heterogeneity = 0.366) and 1.61 (95% CI 1.17-2.21, p = 0.003, p for heterogeneity = 0.616), respectively. For both SNPs, while smoking was significantly less frequent in patients than controls in AA homozygotes, this inverse association disappeared in G allele carriers. CONCLUSIONS We identified and replicated suggestive gene-by-smoking interactions in PD. The inverse association of smoking with PD was less pronounced in carriers of minor alleles of both RXRA-rs4240705 and SLC17A6-rs1900586. These findings may help identify biological pathways involved in the inverse association between smoking and PD.
Collapse
Affiliation(s)
- Pei-Chen Lee
- From the Department of Health Care Management (P.-C.L.), College of Health Technology, National Taipei University of Nursing and Health Sciences, Taiwan; Biostatistics, Biomathematics, Pharmacoepidemiology and Infectious Diseases (I.A.), INSERM, UVSQ, Institut Pasteur, Université Paris-Saclay, Villejuif; Assistance-Publique-Hôpitaux de Paris (M.-A.L.), Hôpital Européen Georges Pompidou, Biochimie, Pharmacogénétique et Oncologie Moléculaire; INSERM UMR-S 1147 (C.M.), CRB EPIGENETEC, Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Departments of Epidemiology (K.C.P., B.R.) and Environmental Health (J.M.B., B.R.), Fielding School of Public Health, and Department of Neurology (J.M.B., B.R.), Geffen School of Medicine, University of California at Los Angeles; and Université Paris-Saclay (A.E.), Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
| | - Ismaïl Ahmed
- From the Department of Health Care Management (P.-C.L.), College of Health Technology, National Taipei University of Nursing and Health Sciences, Taiwan; Biostatistics, Biomathematics, Pharmacoepidemiology and Infectious Diseases (I.A.), INSERM, UVSQ, Institut Pasteur, Université Paris-Saclay, Villejuif; Assistance-Publique-Hôpitaux de Paris (M.-A.L.), Hôpital Européen Georges Pompidou, Biochimie, Pharmacogénétique et Oncologie Moléculaire; INSERM UMR-S 1147 (C.M.), CRB EPIGENETEC, Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Departments of Epidemiology (K.C.P., B.R.) and Environmental Health (J.M.B., B.R.), Fielding School of Public Health, and Department of Neurology (J.M.B., B.R.), Geffen School of Medicine, University of California at Los Angeles; and Université Paris-Saclay (A.E.), Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
| | - Marie-Anne Loriot
- From the Department of Health Care Management (P.-C.L.), College of Health Technology, National Taipei University of Nursing and Health Sciences, Taiwan; Biostatistics, Biomathematics, Pharmacoepidemiology and Infectious Diseases (I.A.), INSERM, UVSQ, Institut Pasteur, Université Paris-Saclay, Villejuif; Assistance-Publique-Hôpitaux de Paris (M.-A.L.), Hôpital Européen Georges Pompidou, Biochimie, Pharmacogénétique et Oncologie Moléculaire; INSERM UMR-S 1147 (C.M.), CRB EPIGENETEC, Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Departments of Epidemiology (K.C.P., B.R.) and Environmental Health (J.M.B., B.R.), Fielding School of Public Health, and Department of Neurology (J.M.B., B.R.), Geffen School of Medicine, University of California at Los Angeles; and Université Paris-Saclay (A.E.), Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
| | - Claire Mulot
- From the Department of Health Care Management (P.-C.L.), College of Health Technology, National Taipei University of Nursing and Health Sciences, Taiwan; Biostatistics, Biomathematics, Pharmacoepidemiology and Infectious Diseases (I.A.), INSERM, UVSQ, Institut Pasteur, Université Paris-Saclay, Villejuif; Assistance-Publique-Hôpitaux de Paris (M.-A.L.), Hôpital Européen Georges Pompidou, Biochimie, Pharmacogénétique et Oncologie Moléculaire; INSERM UMR-S 1147 (C.M.), CRB EPIGENETEC, Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Departments of Epidemiology (K.C.P., B.R.) and Environmental Health (J.M.B., B.R.), Fielding School of Public Health, and Department of Neurology (J.M.B., B.R.), Geffen School of Medicine, University of California at Los Angeles; and Université Paris-Saclay (A.E.), Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
| | - Kimberly C Paul
- From the Department of Health Care Management (P.-C.L.), College of Health Technology, National Taipei University of Nursing and Health Sciences, Taiwan; Biostatistics, Biomathematics, Pharmacoepidemiology and Infectious Diseases (I.A.), INSERM, UVSQ, Institut Pasteur, Université Paris-Saclay, Villejuif; Assistance-Publique-Hôpitaux de Paris (M.-A.L.), Hôpital Européen Georges Pompidou, Biochimie, Pharmacogénétique et Oncologie Moléculaire; INSERM UMR-S 1147 (C.M.), CRB EPIGENETEC, Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Departments of Epidemiology (K.C.P., B.R.) and Environmental Health (J.M.B., B.R.), Fielding School of Public Health, and Department of Neurology (J.M.B., B.R.), Geffen School of Medicine, University of California at Los Angeles; and Université Paris-Saclay (A.E.), Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
| | - Jeff M Bronstein
- From the Department of Health Care Management (P.-C.L.), College of Health Technology, National Taipei University of Nursing and Health Sciences, Taiwan; Biostatistics, Biomathematics, Pharmacoepidemiology and Infectious Diseases (I.A.), INSERM, UVSQ, Institut Pasteur, Université Paris-Saclay, Villejuif; Assistance-Publique-Hôpitaux de Paris (M.-A.L.), Hôpital Européen Georges Pompidou, Biochimie, Pharmacogénétique et Oncologie Moléculaire; INSERM UMR-S 1147 (C.M.), CRB EPIGENETEC, Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Departments of Epidemiology (K.C.P., B.R.) and Environmental Health (J.M.B., B.R.), Fielding School of Public Health, and Department of Neurology (J.M.B., B.R.), Geffen School of Medicine, University of California at Los Angeles; and Université Paris-Saclay (A.E.), Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
| | - Beate Ritz
- From the Department of Health Care Management (P.-C.L.), College of Health Technology, National Taipei University of Nursing and Health Sciences, Taiwan; Biostatistics, Biomathematics, Pharmacoepidemiology and Infectious Diseases (I.A.), INSERM, UVSQ, Institut Pasteur, Université Paris-Saclay, Villejuif; Assistance-Publique-Hôpitaux de Paris (M.-A.L.), Hôpital Européen Georges Pompidou, Biochimie, Pharmacogénétique et Oncologie Moléculaire; INSERM UMR-S 1147 (C.M.), CRB EPIGENETEC, Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Departments of Epidemiology (K.C.P., B.R.) and Environmental Health (J.M.B., B.R.), Fielding School of Public Health, and Department of Neurology (J.M.B., B.R.), Geffen School of Medicine, University of California at Los Angeles; and Université Paris-Saclay (A.E.), Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France
| | - Alexis Elbaz
- From the Department of Health Care Management (P.-C.L.), College of Health Technology, National Taipei University of Nursing and Health Sciences, Taiwan; Biostatistics, Biomathematics, Pharmacoepidemiology and Infectious Diseases (I.A.), INSERM, UVSQ, Institut Pasteur, Université Paris-Saclay, Villejuif; Assistance-Publique-Hôpitaux de Paris (M.-A.L.), Hôpital Européen Georges Pompidou, Biochimie, Pharmacogénétique et Oncologie Moléculaire; INSERM UMR-S 1147 (C.M.), CRB EPIGENETEC, Université Paris Descartes, Sorbonne Paris Cité, Paris, France; Departments of Epidemiology (K.C.P., B.R.) and Environmental Health (J.M.B., B.R.), Fielding School of Public Health, and Department of Neurology (J.M.B., B.R.), Geffen School of Medicine, University of California at Los Angeles; and Université Paris-Saclay (A.E.), Université Paris-Sud, UVSQ, CESP, INSERM, Villejuif, France.
| |
Collapse
|
21
|
Mohammed MM, Sallam AED, Hussein AA, Ibrahim ZN. β-carotene ameliorates CUS-induced circadian alternations of locomotor activity and melatonin patterns in rats. BIOL RHYTHM RES 2017. [DOI: 10.1080/09291016.2017.1350441] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Affiliation(s)
| | - Alaa El-Din Sallam
- Faculty of Science, Zoology Department, Suez Canal University, Ismailia, Egypt
| | - Aida A. Hussein
- Faculty of Science, Zoology Department, Suez University, Suez, Egypt
| | - Zohour N. Ibrahim
- Faculty of Science, Zoology Department, Suez Canal University, Ismailia, Egypt
| |
Collapse
|
22
|
Yu SJ, Airavaara M, Wu KJ, Harvey BK, Liu HS, Yang Y, Zacharek A, Chen J, Wang Y. 9-cis retinoic acid induces neurorepair in stroke brain. Sci Rep 2017; 7:4512. [PMID: 28674431 PMCID: PMC5495771 DOI: 10.1038/s41598-017-04048-2] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2016] [Accepted: 05/09/2017] [Indexed: 11/09/2022] Open
Abstract
The purpose of this study was to examine the neurorestorative effect of delayed 9 cis retinoic acid (9cRA) treatment for stroke. Adult male rats received a 90-min right distal middle cerebral artery occlusion (dMCAo). Animals were separated into two groups with similar infarction sizes, based on magnetic resonance imaging on day 2 after dMCAo. 9cRA or vehicle was given via an intranasal route daily starting from day 3. Stroke rats receiving 9cRA post-treatment showed an increase in brain 9cRA levels and greater recovery in motor function. 9cRA enhanced the proliferation of bromodeoxyuridine (+) cells in the subventricular zone (SVZ) and lesioned cortex in the stroke brain. Using subventricular neurosphere and matrigel cultures, we demonstrated that proliferation and migration of SVZ neuroprogenitor cells were enhanced by 9cRA. Our data support a delayed and non-invasive drug therapy for stroke. Intranasal 9cRA can facilitate the functional recovery and endogenous repair in the ischemic brain.
Collapse
Affiliation(s)
- Seong-Jin Yu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Mikko Airavaara
- Institute of Biotechnology, Viikki Biocenter, University of Helsinki, Helsinki, Finland
| | - Kuo-Jen Wu
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan
| | - Brandon K Harvey
- Intramural Research Program, National Institute on Drug Abuse, NIH, USA
| | - H S Liu
- Intramural Research Program, National Institute on Drug Abuse, NIH, USA
| | - Yihong Yang
- Intramural Research Program, National Institute on Drug Abuse, NIH, USA
| | | | - Jieli Chen
- Neurology, Henry Ford Hospital, Detroit, MI, USA
| | - Yun Wang
- Center for Neuropsychiatric Research, National Health Research Institutes, Zhunan, Taiwan.
| |
Collapse
|
23
|
Requejo-Aguilar R, Bolaños JP. Mitochondrial control of cell bioenergetics in Parkinson's disease. Free Radic Biol Med 2016; 100:123-137. [PMID: 27091692 PMCID: PMC5065935 DOI: 10.1016/j.freeradbiomed.2016.04.012] [Citation(s) in RCA: 51] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Revised: 04/13/2016] [Accepted: 04/14/2016] [Indexed: 12/15/2022]
Abstract
Parkinson disease (PD) is a neurodegenerative disorder characterized by a selective loss of dopaminergic neurons in the substantia nigra. The earliest biochemical signs of the disease involve failure in mitochondrial-endoplasmic reticulum cross talk and lysosomal function, mitochondrial electron chain impairment, mitochondrial dynamics alterations, and calcium and iron homeostasis abnormalities. These changes are associated with increased mitochondrial reactive oxygen species (mROS) and energy deficiency. Recently, it has been reported that, as an attempt to compensate for the mitochondrial dysfunction, neurons invoke glycolysis as a low-efficient mode of energy production in models of PD. Here, we review how mitochondria orchestrate the maintenance of cellular energetic status in PD, with special focus on the switch from oxidative phosphorylation to glycolysis, as well as the implication of endoplasmic reticulum and lysosomes in the control of bioenergetics.
Collapse
Affiliation(s)
- Raquel Requejo-Aguilar
- Department of Biochemistry and Molecular Biology, University of Cordoba, Institute Maimonides of Biomedical Investigation of Cordoba (IMIBIC), Cordoba, Spain
| | - Juan P Bolaños
- Institute of Functional Biology and Genomics (IBFG), University of Salamanca-CSIC, Zacarias Gonzalez, 2, 37007 Salamanca, Spain.
| |
Collapse
|
24
|
Nurr1 and Retinoid X Receptor Ligands Stimulate Ret Signaling in Dopamine Neurons and Can Alleviate α-Synuclein Disrupted Gene Expression. J Neurosci 2016; 35:14370-85. [PMID: 26490873 DOI: 10.1523/jneurosci.1155-15.2015] [Citation(s) in RCA: 44] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
UNLABELLED α-synuclein, a protein enriched in Lewy bodies and highly implicated in neurotoxicity in Parkinson's disease, is distributed both at nerve terminals and in the cell nucleus. Here we show that a nuclear derivative of α-synuclein induces more pronounced changes at the gene expression level in mouse primary dopamine (DA) neurons compared to a derivative that is excluded from the nucleus. Moreover, by RNA sequencing we analyzed the extent of genome-wide effects on gene expression resulting from expression of human α-synuclein in primary mouse DA neurons. The results implicated the transcription factor Nurr1 as a key dysregulated target of α-synuclein toxicity. Forced Nurr1 expression restored the expression of hundreds of dysregulated genes in primary DA neurons expressing α-synuclein, and therefore prompted us to test the possibility that Nurr1 can be pharmacologically targeted by bexarotene, a ligand for the retinoid X receptor that forms heterodimers with Nurr1. Although our data demonstrated that bexarotene was ineffective in neuroprotection in rats in vivo, the results revealed that bexarotene has the capacity to coregulate subsets of Nurr1 target genes including the receptor tyrosine kinase subunit Ret. Moreover, bexarotene was able to restore dysfunctional Ret-dependent neurotrophic signaling in α-synuclein-overexpressing mouse DA neurons. These data highlight the role of the Nurr1-Ret signaling pathway as a target of α-synuclein toxicity and suggest that retinoid X receptor ligands with appropriate pharmacological properties could have therapeutic potential in Parkinson's disease. SIGNIFICANCE STATEMENT How α-synuclein, a protein enriched in Lewy bodies in Parkinson's disease, is causing neuropathology in dopamine neurons remains unclear. This study elucidated how α-synuclein is influencing gene expression and how Nurr1, a transcription factor known to protect dopamine neurons against α-synuclein toxicity, can counteract these effects. Moreover, given the protective role of Nurr1, this study also investigated how Nurr1 could be pharmacologically targeted via bexarotene, a ligand of Nurr1's heterodimerization partner retinoid X receptor (RXR). The results showed that RXR ligands could increase neurotrophic signaling, but provided a mixed picture of its potential in a Parkinson's disease rat model in vivo. However, this study clearly emphasized Nurr1's neuroprotective role and indicated that other RXR ligands could have therapeutic potential in Parkinson's disease.
Collapse
|
25
|
Lone AM, Dar NJ, Hamid A, Shah WA, Ahmad M, Bhat BA. Promise of Retinoic Acid-Triazolyl Derivatives in Promoting Differentiation of Neuroblastoma Cells. ACS Chem Neurosci 2016; 7:82-9. [PMID: 26551203 DOI: 10.1021/acschemneuro.5b00267] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Retinoic acid induces differentiation in various types of cells including skeletal myoblasts and neuroblasts and maintains differentiation of epithelial cells. The present study demonstrates synthesis and screening of a library of retinoic acid-triazolyl derivatives for their differentiation potential on neuroblastoma cells. Click chemistry approach using copper(I)-catalyzed azide-alkyne cycloaddition was adopted for the preparation of these derivatives. The neurite outgrowth promoting potential of retinoic acid-triazolyl derivatives was studied on neuroblastoma cells. Morphological examination revealed that compounds 8a, 8e, 8f, and 8k, among the various derivatives screened, exhibited promising neurite-outgrowth inducing activity at a concentration of 10 μM compared to undifferentiated and retinoic acid treated cells. Further on, to confirm this differentiation potential of these compounds, neuroblastoma cells were probed for expression of neuronal markers such as NF-H and NeuN. The results revealed a marked increase in the NF-H and NeuN protein expression when treated with 8a, 8e, 8f, and 8k compared to undifferentiated and retinoic acid treated cells. Thus, these compounds could act as potential leads in inducing neuronal differentiation for future studies.
Collapse
Affiliation(s)
| | - Nawab John Dar
- Academy
of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Abid Hamid
- Cancer
Pharmacology Division, CSIR-Indian Institute of Integrative Medicine, Jammu 180001, India
- Academy
of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Wajaht Amin Shah
- Department
of Chemistry, University of Kashmir, Srinagar 190006, India
| | - Muzamil Ahmad
- Academy
of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| | - Bilal A. Bhat
- Academy
of Scientific and Innovative Research, CSIR-Indian Institute of Integrative Medicine, Canal Road, Jammu 180001, India
| |
Collapse
|
26
|
Wang F, Du M, Wang R, Zhou J, Zhang W, Li H. Molecular mechanism of Hoxd13-mediated congenital malformations in rat embryos. INTERNATIONAL JOURNAL OF CLINICAL AND EXPERIMENTAL PATHOLOGY 2015; 8:15591-15598. [PMID: 26884828 PMCID: PMC4730041] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Subscribe] [Scholar Register] [Received: 07/29/2015] [Accepted: 08/28/2015] [Indexed: 06/05/2023]
Abstract
OBJECTIVE To investigate the molecular mechanism of Hoxd13-mediated congenital malformations in rat embryos. METHODS SD female rats were mated with male rats in a 1:1 mating scheme. Thirty pregnant female rats were randomly divided into three groups: the control group receiving a normal diet, the model group receiving a vitamin A-deficient diet, and the treatment group receiving a vitamin A-deficient diet supplemented with pcDNA-Hoxd13. The expression of Hoxd13 mRNA and protein in normal embryonic tissue and congenital malformations was determined by RT-PCR and Western blot analysis. At day 20, rats were dissected, and the fetal weight, body and tail length, and the number of live births, absorbed fetus, and stillbirth in each group were recorded. Wnt and Slim1 expression was detected by RT-PCR and Western blot analysis. β-catenin and c-myc expression was also quantified by Western blot analysis. RESULTS The expression of Hoxd13 mRNA and protein in congenital malformations was significantly lower compared with normal embryonic tissue (P<0.01). The administration of exogenous Hoxd13 in the treatment group markedly increased the fetal weight, body and tail length (P<0.05), improved the embryonic survival rate, and reduced the embryonic resorption rate and stillbirth rate (P<0.05). Exogenous Hoxd13 markedly promoted the expression of Wnt2, Wnt5a, Wnt7b and Slim1 protein and mRNA (P<0.01), and the expression of β-catenin and c-myc protein in congenital malformations (P<0.01). CONCLUSION Hoxd13 expression was decreased in rat embryos with congenital malformations. The administration of exogenous Hoxd13 alleviated fetal malformation probably through stimulation of Slim1 expression and Wnt/β-catenin signaling pathway.
Collapse
Affiliation(s)
- Fenglan Wang
- Department of Ultrasonography, Maternal and Children Health Hospital of Tangshan CityChina
| | - Mingzhen Du
- Department of Ultrasonography, Maternal and Children Health Hospital of Tangshan CityChina
| | - Ruiling Wang
- Department of Ultrasonography, Maternal and Children Health Hospital of Tangshan CityChina
| | - Juekun Zhou
- Department of Ultrasonography, Maternal and Children Health Hospital of Tangshan CityChina
| | - Wei Zhang
- Department of Ultrasonography, Maternal and Children Health Hospital of Tangshan CityChina
| | - Huixue Li
- Department of Ultrasonography, The Affiliated Hospital of North China University of Science and TechnologyChina
| |
Collapse
|
27
|
Wevers NR, de Vries HE. Morphogens and blood-brain barrier function in health and disease. Tissue Barriers 2015; 4:e1090524. [PMID: 27141417 DOI: 10.1080/21688370.2015.1090524] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/18/2015] [Revised: 08/26/2015] [Accepted: 08/30/2015] [Indexed: 12/22/2022] Open
Abstract
The microvasculature of the brain forms a protective blood-brain barrier (BBB) that ensures a homeostatic environment for the central nervous system (CNS), which is essential for optimal brain functioning. The barrier properties of the brain endothelial cells are maintained by cells surrounding the capillaries, such as astrocytes and pericytes. Together with the endothelium and a basement membrane, these supporting cells form the neurovascular unit (NVU). Accumulating evidence indicates that the supporting cells of the NVU release a wide variety of soluble factors that induce and control barrier properties in a concentration-dependent manner. The current review provides a comprehensive overview of how such factors, called morphogens, influence BBB integrity and functioning. Since impaired BBB function is apparent in numerous CNS disorders and is often associated with disease severity, we also discuss the potential therapeutic value of these morphogens, as they may represent promising therapies for a wide variety of CNS disorders.
Collapse
Affiliation(s)
| | - Helga E de Vries
- Department of Molecular Cell Biology and Immunology; Neuroscience Campus Amsterdam, VU University Medical Center ; Amsterdam, The Netherlands
| |
Collapse
|
28
|
Alquézar C, Barrio E, Esteras N, de la Encarnación A, Bartolomé F, Molina JA, Martín-Requero Á. Targeting cyclin D3/CDK6 activity for treatment of Parkinson's disease. J Neurochem 2015; 133:886-97. [PMID: 25689470 DOI: 10.1111/jnc.13070] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2014] [Revised: 02/10/2015] [Accepted: 02/10/2015] [Indexed: 01/11/2023]
Abstract
At present, treatment for Parkinson's disease (PD) is only symptomatic; therefore, it is important to identify new targets tackling the molecular causes of the disease. We previously found that lymphoblasts from sporadic PD patients display increased activity of the cyclin D3/CDK6/pRb pathway and higher proliferation than control cells. These features were considered systemic manifestations of the disease, as aberrant activation of the cell cycle is involved in neuronal apoptosis. The main goal of this work was to elucidate whether the inhibition of cyclin D3/CDK6-associated kinase activity could be useful in PD treatment. For this purpose, we investigated the effects of two histone deacetylase (HDAC) inhibitors, suberoylanilide hydroxamic (SAHA) acid and sodium butyrate (NaB), and the m-TOR inhibitor rapamycin on cell viability and cyclin D3/CDK6 activity. Moreover, the potential neuroprotective action of these drugs was evaluated in 6-hydroxy-dopamine (6-OHDA) treated dopaminergic SH-SY5Y cells and primary rat mesencephalic cultures. Here, we report that both compounds normalized the proliferative activity of PD lymphoblasts and reduced the 6-OHDA-induced cell death in neuronal cells by preventing the over-activation of the cyclin D3/CDK6/pRb cascade. Considering that these drugs are already used in clinic for treatment of other diseases with good tolerance, it is plausible that they may serve as novel therapeutic drugs for PD. We report here that peripheral cells from Parkinson's disease (PD) patients show an enhanced proliferative activity due to the activation of cyclin D3/CDK6-mediated phosphorylation of retinoblastoma protein (pRb). Treatment of PD lymphoblasts with inhibitors of histone deacetylases like suberoylanilide hydroxamic acid (SAHA) and sodium butyrate (NaB), or with rapamycin, inhibitor of mechanistic target of rapamycin (mTOR) normalized the proliferation of PD lymphoblasts by preventing the over-activation of the cyclin D3/CDK6/pRb cascade. These drugs were shown to have neuroprotective effects in both human neuroblastoma SH-SY5Y cells and primary rat mid-brain dopaminergic neuronal cultures toxicity induced by 6-hidroxydopamine. Considering that these drugs are already used in clinic for treatment of other diseases with good tolerance, it seems reasonable to believe that the repositioning of these drugs toward PD holds promise as a novel therapeutic strategy.
Collapse
Affiliation(s)
- Carolina Alquézar
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| | - Estíbaliz Barrio
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - Noemí Esteras
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - Ana de la Encarnación
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain
| | - Fernando Bartolomé
- Neuroscience Laboratory, Research Institute, Hospital Doce de Octubre, Madrid, Spain
| | - José A Molina
- Department of Neurology, Hospital Doce de Octubre, Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Neurodegenerativas (CIBERNED), Madrid, Spain
| | - Ángeles Martín-Requero
- Department of Cellular and Molecular Medicine, Centro de Investigaciones Biológicas (CSIC), Madrid, Spain.,Centro de Investigación Biomédica en Red de Enfermedades Raras (CIBERER), Madrid, Spain
| |
Collapse
|
29
|
Kandratavicius L, Balista PA, Wolf DC, Abrao J, Evora PR, Rodrigues AJ, Chaves C, Maia-de-Oliveira JP, Leite JP, Dursun SM, Baker GB, Guimaraes FS, Hallak JEC. Effects of nitric oxide-related compounds in the acute ketamine animal model of schizophrenia. BMC Neurosci 2015; 16:9. [PMID: 25887360 PMCID: PMC4354998 DOI: 10.1186/s12868-015-0149-3] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2014] [Accepted: 02/25/2015] [Indexed: 01/22/2023] Open
Abstract
Background Better treatments for schizophrenia are urgently needed. The therapeutic use of the nitric oxide (NO)-donor sodium nitroprusside (SNP) in patients with schizophrenia has shown promising results. The role of NO in schizophrenia is still unclear, and NO modulation is unexplored in ketamine (KET) animal models to date. In the present study, we compared the behavioral effects of pre- and post-treatment with SNP, glyceryl trinitrate (GTN), and methylene blue (MB) in the acute KET animal model of schizophrenia. The present study was designed to test whether acute SNP, GTN, and MB treatment taken after (therapeutic effect) or before (preventive effect) a single KET injection would influence the behavior of rats in the sucrose preference test, object recognition task and open field. Results The results showed that KET induced cognitive deficits and hyperlocomotion. Long- term memory improvement was seen with the therapeutic GTN and SNP treatment, but not with the preventive one. MB pretreatment resulted in long-term memory recovery. GTN pre-, but not post-treatment, tended to increase vertical and horizontal activity in the KET model. Therapeutic and preventive SNP treatment consistently decreased KET-induced hyperlocomotion. Conclusion NO donors – especially SNP – are promising new pharmacological candidates in the treatment of schizophrenia. In addition, we showed that the potential impact of NO-related compounds on KET-induced behavioral changes may depend on the temporal window of drug administration.
Collapse
Affiliation(s)
- Ludmyla Kandratavicius
- Department of Neurosciences and Behavior, Ribeirao Preto School of Medicine, University of Sao Paulo, Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, Brazil. .,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), USP, Ribeirao Preto, Brazil.
| | - Priscila Alves Balista
- Department of Neurosciences and Behavior, Ribeirao Preto School of Medicine, University of Sao Paulo, Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, Brazil.
| | - Daniele Cristina Wolf
- Department of Neurosciences and Behavior, Ribeirao Preto School of Medicine, University of Sao Paulo, Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, Brazil.
| | - Joao Abrao
- Department of Biomechanics, Ribeirao Preto School of Medicine, Medicine and Rehabilitation, USP, Ribeirao Preto, Brazil.
| | - Paulo Roberto Evora
- Department of Surgery and Anatomy, Ribeirao Preto School of Medicine, USP, Ribeirao Preto, Brazil.
| | - Alfredo Jose Rodrigues
- Department of Surgery and Anatomy, Ribeirao Preto School of Medicine, USP, Ribeirao Preto, Brazil.
| | - Cristiano Chaves
- Department of Neurosciences and Behavior, Ribeirao Preto School of Medicine, University of Sao Paulo, Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, Brazil.
| | | | - Joao Pereira Leite
- Department of Neurosciences and Behavior, Ribeirao Preto School of Medicine, University of Sao Paulo, Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, Brazil. .,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), USP, Ribeirao Preto, Brazil.
| | - Serdar Murat Dursun
- Department of Psychiatry (NRU), University of Alberta, Edmonton, Alberta, Canada.
| | - Glen Bryan Baker
- Department of Psychiatry (NRU), University of Alberta, Edmonton, Alberta, Canada.
| | | | - Jaime Eduardo Cecilio Hallak
- Department of Neurosciences and Behavior, Ribeirao Preto School of Medicine, University of Sao Paulo, Av Bandeirantes 3900, CEP 14049-900, Ribeirao Preto, Brazil. .,Center for Interdisciplinary Research on Applied Neurosciences (NAPNA), USP, Ribeirao Preto, Brazil. .,National Institute of Science and Technology in Translational Medicine (INCT-TM - CNPq), Ribeirao Preto, Brazil.
| |
Collapse
|
30
|
Esteves M, Cristóvão AC, Saraiva T, Rocha SM, Baltazar G, Ferreira L, Bernardino L. Retinoic acid-loaded polymeric nanoparticles induce neuroprotection in a mouse model for Parkinson's disease. Front Aging Neurosci 2015; 7:20. [PMID: 25798108 PMCID: PMC4351630 DOI: 10.3389/fnagi.2015.00020] [Citation(s) in RCA: 52] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 02/18/2015] [Indexed: 11/13/2022] Open
Abstract
Retinoic acid (RA) plays an important role in the commitment, maturation and survival of neural cells. Recently, RA was pointed as a therapeutic option for some neurodegenerative diseases, including Parkinson's disease (PD). The administration of RA has been defying, and in this sense we have previously developed novel RA-loaded polymeric nanoparticles (RA-NPs) that ensure the efficient intracellular transport and controlled release of RA. Herein, we show that nanoformulation as an efficient neuroprotective effect on dopaminergic (DA) neurons in the 1-methyl-4-phenyl-1, 2, 3, 6-tetrahydropyridine (MPTP) induced mouse model for PD. The results showed that the RA-NPs administration induced a significant reduction of DA neuron loss in the substantia nigra (SN) as well as their neuronal fiber/axonal innervations in the striatum. Furthermore, we observed an increase in the expression levels of the transcription factors Pitx3 and Nurr1 induced by RA-NPs, showing its supportive effect on the development and functional maintenance of DA neurons in PD. This is the first study showing that RA-NPs can be an innovative strategy to halt the progression of PD pathogenesis, suggesting that this nanoformulation could be of particular interest for the development of new approaches for PD therapeutics.
Collapse
Affiliation(s)
- Marta Esteves
- Faculty of Health Sciences, Health Sciences Research Centre, University of Beira Interior Covilhã, Portugal
| | - Ana C Cristóvão
- Faculty of Health Sciences, Health Sciences Research Centre, University of Beira Interior Covilhã, Portugal
| | - Tatiana Saraiva
- Faculty of Health Sciences, Health Sciences Research Centre, University of Beira Interior Covilhã, Portugal
| | - Sandra M Rocha
- Faculty of Health Sciences, Health Sciences Research Centre, University of Beira Interior Covilhã, Portugal
| | - Graça Baltazar
- Faculty of Health Sciences, Health Sciences Research Centre, University of Beira Interior Covilhã, Portugal
| | - Lino Ferreira
- Center for Neuroscience and Cell Biology, University of Coimbra Coimbra, Portugal ; Biocant - Center of Innovation in Biotechnology Cantanhede, Portugal
| | - Liliana Bernardino
- Faculty of Health Sciences, Health Sciences Research Centre, University of Beira Interior Covilhã, Portugal
| |
Collapse
|
31
|
Szutowicz A, Bielarczyk H, Jankowska-Kulawy A, Ronowska A, Pawełczyk T. Retinoic acid as a therapeutic option in Alzheimer's disease: a focus on cholinergic restoration. Expert Rev Neurother 2015; 15:239-49. [PMID: 25683350 DOI: 10.1586/14737175.2015.1008456] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023]
Abstract
Retinoic acid is a potent cell differentiating factor, which through its nuclear receptors affects a vast range of promoter sites in brain neuronal and glial cells in every step of embryonic and postnatal life. Its capacities, facilitating maturation of neurotransmitter phenotype in different groups of neurons, pave the way for its application as a potential therapeutic agent in neurodegenerative diseases including Alzheimer's disease. Retinoic acid was found to exert particularly strong enhancing effects on acetylcholine transmitter functions in brain cholinergic neurons, loss of which is tightly linked to the development of cognitive and memory deficits in course of different cholinergic encephalopathies. Here, we review cholinotrophic properties of retinoic acid and its derivatives, which may justify their application in the management of Alzheimer's disease and the related neurodegenerative conditions.
Collapse
Affiliation(s)
- Andrzej Szutowicz
- Department of Laboratory Medicine, Medical University of Gdańsk, Gdańsk, Poland
| | | | | | | | | |
Collapse
|
32
|
Haybaeck J, Postruznik M, Miller CL, Dulay JR, Llenos IC, Weis S. Increased expression of retinoic acid-induced gene 1 in the dorsolateral prefrontal cortex in schizophrenia, bipolar disorder, and major depression. Neuropsychiatr Dis Treat 2015; 11:279-89. [PMID: 25678793 PMCID: PMC4322876 DOI: 10.2147/ndt.s72536] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
BACKGROUND Retinoids regulate gene expression in different cells and tissues at the transcriptional level. Retinoic acid transcriptionally regulates downstream regulatory molecules, including enzymes, transcription factors, cytokines, and cytokine receptors. Animal models indicate an involvement of retinoid signaling pathways in the regulation of synaptic plasticity and learning, especially in the hippocampus. Retinoic acid-inducible or induced gene 1 (RAI-1) is induced during neuronal differentiation, and was associated with the severity of the phenotype and response to medication in schizophrenic patients. METHODS In the present study, we used immunohistochemistry to investigate the expression of RAI-1 in 60 brains from the Stanley Neuropathology Consortium (15 cases each from controls and from patients with schizophrenia, bipolar disorder, and major depression). Rating scores for density and intensity were determined in the dorsolateral prefrontal cortex. RESULTS All four groups showed high interindividual variation. RAI-1-positive cells were identified as neurons and astrocytes. Significantly increased intensities in cortical neurons were noted in all three major psychiatric groups compared with controls. The density of RAI-1-positive neurons was increased (P=0.06) in schizophrenia and bipolar disorder. In bipolar disorder, RAI-1-positive astrocytes in gray matter showed a significantly increased intensity and compound value. Thus, a significant increase in the parameters measured was found in schizophrenia, bipolar disorder, and major depression. CONCLUSION Our study shows a significant increase in expression of RAI-1 in the brains from patients with schizophrenia, bipolar disorder, or major depression. The increased expression might reflect altered signaling pathways, like that for retinoic acid. The underlying mechanisms leading to the increased expression and its functional consequences are so far unknown, and remain to be investigated in future studies.
Collapse
Affiliation(s)
- Johannes Haybaeck
- Department of Neuropathology, Institute of Pathology, Medical University Graz, Graz, Austria
| | - Magdalena Postruznik
- Department of Neuropathology, Institute of Pathology, Medical University Graz, Graz, Austria
| | | | - Jeannette R Dulay
- Laboratory of Brain Research and Neuropathology, Departments of Psychiatry and Pathology, Uniformed Services University of the Health Sciences, and Stanley Medical Research Institute, Bethesda, MD, USA
| | - Ida C Llenos
- Laboratory of Brain Research and Neuropathology, Departments of Psychiatry and Pathology, Uniformed Services University of the Health Sciences, and Stanley Medical Research Institute, Bethesda, MD, USA ; Laboratory of Neuropathology, Department of Pathology and Neuropathology, State Neuropsychiatric Hospital Wagner-Jauregg, Medical School, Johannes Kepler University, Linz, Austria
| | - Serge Weis
- Laboratory of Brain Research and Neuropathology, Departments of Psychiatry and Pathology, Uniformed Services University of the Health Sciences, and Stanley Medical Research Institute, Bethesda, MD, USA ; Laboratory of Neuropathology, Department of Pathology and Neuropathology, State Neuropsychiatric Hospital Wagner-Jauregg, Medical School, Johannes Kepler University, Linz, Austria
| |
Collapse
|
33
|
Reiner DJ, Yu SJ, Shen H, He Y, Bae E, Wang Y. 9-Cis retinoic acid protects against methamphetamine-induced neurotoxicity in nigrostriatal dopamine neurons. Neurotox Res 2013; 25:248-61. [PMID: 23884514 DOI: 10.1007/s12640-013-9413-4] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 07/11/2013] [Accepted: 07/13/2013] [Indexed: 01/03/2023]
Abstract
Methamphetamine (MA) is a drug of abuse as well as a dopaminergic neurotoxin. 9-Cis retinoic acid (9cRA), a biologically active derivative of vitamin A, has protective effects against damage caused by H(2)O(2) and oxygen-glucose deprivation in vitro as well as infarction and terminal deoxynucleotidyl transferase-mediated dNTP nick-end labeling (TUNEL) labeling in ischemic brain. The purpose of this study was to examine if there was a protective role for 9cRA against MA toxicity in nigrostriatal dopaminergic neurons. Primary dopaminergic neurons, prepared from rat embryonic ventral mesencephalic tissue, were treated with MA. High doses of MA decreased tyrosine hydroxylase (TH) immunoreactivity while increasing TUNEL labeling. These toxicities were significantly reduced by 9cRA. 9cRA also inhibited the export of Nur77 from nucleus to cytosol, a response that activates apoptosis. The interaction of 9cRA and MA in vivo was next examined in adult rats. 9cRA was delivered intracerebroventricularly; MA was given (5 mg/kg, 4×) one day later. Locomotor behavior was measured 2 days after surgery for a period of 48 h. High doses of MA significantly reduced locomotor activity and TH immunoreactivity in striatum. Administration of 9cRA antagonized these changes. Previous studies have shown that 9cRA can induce bone morphogenetic protein-7 (BMP7) expression and that administration of BMP7 attenuates MA toxicity. We demonstrated that MA treatment significantly reduced BMP7 mRNA expression in nigra. Noggin (a BMP antagonist) antagonized 9cRA-induced behavioral recovery and 9cRA-induced normalization of striatal TH levels. Our data suggest that 9cRA has a protective effect against MA-mediated neurodegeneration in dopaminergic neurons via upregulation of BMP.
Collapse
Affiliation(s)
- David J Reiner
- Neural Protection and Regeneration Section, Intramural Research Program, National Institute on Drug Abuse, NIH, Baltimore, MD, 21224, USA
| | | | | | | | | | | |
Collapse
|