1
|
Yedke NG, Arthur R, Kumar P. Bacillus calmette gaurine vaccine ameliorates the neurotoxicity of quinolinic acid in rats via the modulation of antioxidant, inflammatory and apoptotic markers. J Chem Neuroanat 2023; 131:102287. [PMID: 37172828 DOI: 10.1016/j.jchemneu.2023.102287] [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: 01/25/2023] [Revised: 04/06/2023] [Accepted: 05/09/2023] [Indexed: 05/15/2023]
Abstract
A mutation in the Huntingtin gene causes 'Huntington's disease, which presents as a motor and behavioral impairment. Due to the limited drug therapy for this disease, scientists are constantly searching for newer and alternative drugs that may either retard or prevent the progress of the disease. This study aims to explore the neuroprotective potential of Bacillus Calmette Gaurine (BCG) vaccine against quinolinic acid-induced (QA) neurotoxicity in rats. QA (200 nmol/2 µl, i.s) was injected bilaterally into the rat striatum, after which a single dose of BCG (2 × 10^7, cfu) was given to the rats. Animals were assessed for behavioral parameters on the 14th and 21st days. On the 22nd day, animals were sacrificed, brains were harvested, and striatum was separated to evaluate biochemical, inflammatory, and apoptotic mediators. Histopathological studies were performed using Hematoxyline and Eosin staining to assess neuronal morphology. BCG treatment reversed motor abnormalities, reduced oxidative stress and neuroinflammatory markers, apoptotic mediators and striatal lesions induced by QA treatment. In conclusion, treat' 'ing rats with BCG vaccine (2 × 10^7, cfu) mitigated the quinolinic acid-induced Huntington's disease-like symptoms. Hence, BCG vaccine (2 ×10^7, cfu) could be used as an adjuvant in managing HD.
Collapse
Affiliation(s)
- Narhari Gangaram Yedke
- Department of Pharmaceutical Sciences and Technology, Maharaja Ranjit Singh Punjab Technical University, Bathinda 151001, Punjab, India; Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Richmond Arthur
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India
| | - Puneet Kumar
- Department of Pharmacology, Central University of Punjab, Ghudda, Bathinda, India.
| |
Collapse
|
2
|
Vázquez-González D, Corona JC. Pioglitazone enhances brain mitochondrial biogenesis and phase II detoxification capacity in neonatal rats with 6-OHDA-induced unilateral striatal lesions. Front Neurosci 2023; 17:1186520. [PMID: 37575308 PMCID: PMC10416244 DOI: 10.3389/fnins.2023.1186520] [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: 03/16/2023] [Accepted: 07/17/2023] [Indexed: 08/15/2023] Open
Abstract
The psychostimulant methylphenidate (MPH) is the first-line pharmacological treatment for attention-deficit/hyperactivity disorder (ADHD), but has numerous adverse side effects. The PPARγ receptor agonist pioglitazone (PIO) is known to improve mitochondrial bioenergetics and antioxidant capacity, both of which may be deficient in ADHD, suggesting utility as an adjunct therapy. Here, we assessed the effects of PIO on ADHD-like symptoms, mitochondrial biogenesis and antioxidant pathways in multiple brain regions of neonate rats with unilateral striatal lesions induced by 6-hydroxydopamine (6-OHDA) as an experimental ADHD model. Unilateral striatal injection of 6-OHDA reduced ipsilateral dopaminergic innervation by 33% and increased locomotor activity. This locomotor hyperactivity was not altered by PIO treatment for 14 days. However, PIO increased the expression of proteins contributing to mitochondrial biogenesis in the striatum, hippocampus, cerebellum and prefrontal cortex of 6-OHDA-lesioned rats. In addition, PIO treatment enhanced the expression of the phase II transcription factor Nrf2 in the striatum, prefrontal cortex and cerebellum. In contrast, no change in the antioxidant enzyme catalase was observed in any of the brain regions analyzed. Thus, PIO may improve mitochondrial biogenesis and phase 2 detoxification in the ADHD brain. Further studies are required to determine if different dose regimens can exert more comprehensive therapeutic effects against ADHD neuropathology and behavior.
Collapse
Affiliation(s)
| | - Juan Carlos Corona
- Laboratory of Neurosciences, Hospital Infantil de México Federico Gómez, Mexico City, Mexico
| |
Collapse
|
3
|
Pizcueta P, Vergara C, Emanuele M, Vilalta A, Rodríguez-Pascau L, Martinell M. Development of PPARγ Agonists for the Treatment of Neuroinflammatory and Neurodegenerative Diseases: Leriglitazone as a Promising Candidate. Int J Mol Sci 2023; 24:ijms24043201. [PMID: 36834611 PMCID: PMC9961553 DOI: 10.3390/ijms24043201] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2022] [Revised: 01/21/2023] [Accepted: 02/01/2023] [Indexed: 02/08/2023] Open
Abstract
Increasing evidence suggests that the peroxisome proliferator-activated receptor γ (PPARγ), a member of the nuclear receptor superfamily, plays an important role in physiological processes in the central nervous system (CNS) and is involved in cellular metabolism and repair. Cellular damage caused by acute brain injury and long-term neurodegenerative disorders is associated with alterations of these metabolic processes leading to mitochondrial dysfunction, oxidative stress, and neuroinflammation. PPARγ agonists have demonstrated the potential to be effective treatments for CNS diseases in preclinical models, but to date, most drugs have failed to show efficacy in clinical trials of neurodegenerative diseases including amyotrophic lateral sclerosis, Parkinson's disease, and Alzheimer's disease. The most likely explanation for this lack of efficacy is the insufficient brain exposure of these PPARγ agonists. Leriglitazone is a novel, blood-brain barrier (BBB)-penetrant PPARγ agonist that is being developed to treat CNS diseases. Here, we review the main roles of PPARγ in physiology and pathophysiology in the CNS, describe the mechanism of action of PPARγ agonists, and discuss the evidence supporting the use of leriglitazone to treat CNS diseases.
Collapse
Affiliation(s)
- Pilar Pizcueta
- Minoryx Therapeutics SL, 08302 Barcelona, Spain
- Correspondence:
| | | | - Marco Emanuele
- Minoryx Therapeutics BE, Gosselies, 6041 Charleroi, Belgium
| | | | | | - Marc Martinell
- Minoryx Therapeutics SL, 08302 Barcelona, Spain
- Minoryx Therapeutics BE, Gosselies, 6041 Charleroi, Belgium
| |
Collapse
|
4
|
Kumar S, Mehan S, Narula AS. Therapeutic modulation of JAK-STAT, mTOR, and PPAR-γ signaling in neurological dysfunctions. J Mol Med (Berl) 2023; 101:9-49. [PMID: 36478124 DOI: 10.1007/s00109-022-02272-6] [Citation(s) in RCA: 12] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Revised: 10/10/2022] [Accepted: 11/11/2022] [Indexed: 12/12/2022]
Abstract
The cytokine-activated Janus kinase (JAK)-signal transducer and activator of transcription (STAT) cascade is a pleiotropic pathway that involves receptor subunit multimerization. The mammalian target of rapamycin (mTOR) is a ubiquitously expressed serine-threonine kinase that perceives and integrates a variety of intracellular and environmental stimuli to regulate essential activities such as cell development and metabolism. Peroxisome proliferator-activated receptor-gamma (PPARγ) is a prototypical metabolic nuclear receptor involved in neural differentiation and axon polarity. The JAK-STAT, mTOR, and PPARγ signaling pathways serve as a highly conserved signaling hub that coordinates neuronal activity and brain development. Additionally, overactivation of JAK/STAT, mTOR, and inhibition of PPARγ signaling have been linked to various neurocomplications, including neuroinflammation, apoptosis, and oxidative stress. Emerging research suggests that even minor disruptions in these cellular and molecular processes can have significant consequences manifested as neurological and neuropsychiatric diseases. Of interest, target modulators have been proven to alleviate neuronal complications associated with acute and chronic neurological deficits. This research-based review explores the therapeutic role of JAK-STAT, mTOR, and PPARγ signaling modulators in preventing neuronal dysfunctions in preclinical and clinical investigations.
Collapse
Affiliation(s)
- Sumit Kumar
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Punjab, Moga, India
| | - Sidharth Mehan
- Division of Neuroscience, Department of Pharmacology, ISF College of Pharmacy, Punjab, Moga, India.
| | - Acharan S Narula
- Narula Research, LLC, 107 Boulder Bluff, Chapel Hill, NC, 27516, USA
| |
Collapse
|
5
|
Mansour RM, El Sayed NS, Ahmed MAE, El-Sahar AE. Addressing Peroxisome Proliferator-Activated Receptor-gamma in 3-Nitropropionic Acid-Induced Striatal Neurotoxicity in Rats. Mol Neurobiol 2022; 59:4368-4383. [PMID: 35553009 PMCID: PMC9167199 DOI: 10.1007/s12035-022-02856-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/27/2022] [Accepted: 04/27/2022] [Indexed: 11/28/2022]
Abstract
Telmisartan (TEL) is an angiotensin II type 1 receptor blocker and a partial activator of peroxisome proliferator-activated receptor-gamma (PPARγ), which regulates inflammatory and apoptotic pathways. Increasing evidence has demonstrated the PPARγ agonistic property of TEL in several brain disorders. This study aims to explore the neuroprotective impact of TEL in 3-nitropropionic acid (3-NP)-induced neurotoxicity in rats. The PPARγ effect of TEL was affirmed by using the PPARγ agonist pioglitazone (PIO), and the antagonist GW9662. 3-NP led to a significant reduction in body weight alongside motor and cognitive functioning. The striata of the 3-NP-treated rats showed energy-deficit, microglia-mediated inflammatory reactions, apoptotic damage as well as histopathological lesions. PIO and TEL improved motor and cognitive perturbations induced by 3-NP, as confirmed by striatal histopathological examination, energy restoration, and neuronal preservation. Both drugs improved mitochondrial biogenesis evidenced by elevated mRNA expression of PPARγ, PGC-1α, and TFAM, alongside increased striatal ATP and SDH. The mitochondrial effect of TEL was beyond PPARγ activation. As well, their anti-inflammatory effect was attributed to suppression of microglial activation, and protein expression of pS536 p65 NF-κB with marked attenuation of striatal inflammatory mediator's release. Anti-inflammatory cytokine IL-10 expression was concurrently increased. TEL effectively participated in neuronal survival as it promoted phosphorylation of Akt/GSK-3β, further increased Bcl-2 expression, and inhibited cleavage of caspase-3. Interestingly, co-treatment with GW9662 partially revoked the beneficial effects of TEL. These findings recommend that TEL improves motor and cognitive performance, while reducing neuronal inflammation and apoptosis in 3-NP-induced neurotoxicity via a PPARγ-dependent mechanism.
Collapse
Affiliation(s)
- Riham M Mansour
- Department of Pharmacology and Toxicology, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), 6Th of October City, Giza, Egypt.
| | - Nesrine S El Sayed
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, Egypt
| | - Maha A E Ahmed
- Department of Pharmacology and Toxicology, College of Pharmaceutical Sciences and Drug Manufacturing, Misr University for Science and Technology (MUST), 6Th of October City, Giza, Egypt
| | - Ayman E El-Sahar
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Cairo University, Kasr El-Aini Street, Cairo, Egypt
| |
Collapse
|
6
|
Plumbagin Alleviates Intracerebroventricular-Quinolinic Acid Induced Depression-like Behavior and Memory Deficits in Wistar Rats. Molecules 2022; 27:molecules27061834. [PMID: 35335195 PMCID: PMC8955906 DOI: 10.3390/molecules27061834] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 03/04/2022] [Accepted: 03/08/2022] [Indexed: 02/01/2023] Open
Abstract
Plumbagin, a hydroxy-1,4-naphthoquinone, confers neuroprotection via antioxidant and anti-inflammatory properties. The present study aimed to assess the effect of plumbagin on behavioral and memory deficits induced by intrahippocampal administration of Quinolinic acid (QA) in male Wistar rats and reveal the associated mechanisms. QA (300 nM/4 μL in Normal saline) was administered i.c.v. in the hippocampus. QA administration caused depression-like behavior (forced swim test and tail suspension tests), anxiety-like behavior (open field test and elevated plus maze), and elevated anhedonia behavior (sucrose preference test). Furthermore, oxidative–nitrosative stress (increased nitrite content and lipid peroxidation with reduction of GSH), inflammation (increased IL-1β), cholinergic dysfunction, and mitochondrial complex (I, II, and IV) dysfunction were observed in the hippocampus region of QA-treated rats as compared to normal controls. Plumbagin (10 and 20 mg/kg; p.o.) treatment for 21 days significantly ameliorated behavioral and memory deficits in QA-administered rats. Moreover, plumbagin treatment restored the GSH level and reduced the MDA and nitrite level in the hippocampus. Furthermore, QA-induced cholinergic dysfunction and mitochondrial impairment were found to be ameliorated by plumbagin treatment. In conclusion, our results suggested that plumbagin offers a neuroprotective potential that could serve as a promising pharmacological approach to mitigate neurobehavioral changes associated with neurodegeneration.
Collapse
|
7
|
Role of Phytoconstituents as PPAR Agonists: Implications for Neurodegenerative Disorders. Biomedicines 2021; 9:biomedicines9121914. [PMID: 34944727 PMCID: PMC8698906 DOI: 10.3390/biomedicines9121914] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/28/2021] [Revised: 12/09/2021] [Accepted: 12/11/2021] [Indexed: 12/16/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPAR-γ, PPAR-α, and PPAR-β/δ) are ligand-dependent nuclear receptors that play a critical role in the regulation of hundreds of genes through their activation. Their expression and targeted activation play an important role in the treatment of a variety of diseases, including neurodegenerative, cardiovascular, diabetes, and cancer. In recent years, several reviews have been published describing the therapeutic potential of PPAR agonists (natural or synthetic) in the disorders listed above; however, no comprehensive report defining the role of naturally derived phytoconstituents as PPAR agonists targeting neurodegenerative diseases has been published. This review will focus on the role of phytoconstituents as PPAR agonists and the relevant preclinical studies and mechanistic insights into their neuroprotective effects. Exemplary research includes flavonoids, fatty acids, cannabinoids, curcumin, genistein, capsaicin, and piperine, all of which have been shown to be PPAR agonists either directly or indirectly. Additionally, a few studies have demonstrated the use of clinical samples in in vitro investigations. The role of the fruit fly Drosophila melanogaster as a potential model for studying neurodegenerative diseases has also been highlighted.
Collapse
|
8
|
Saroj P, Bansal Y, Singh R, Akhtar A, Sodhi RK, Bishnoi M, Sah SP, Kuhad A. Neuroprotective effects of roflumilast against quinolinic acid-induced rat model of Huntington's disease through inhibition of NF-κB mediated neuroinflammatory markers and activation of cAMP/CREB/BDNF signaling pathway. Inflammopharmacology 2021; 29:499-511. [PMID: 33517508 DOI: 10.1007/s10787-020-00787-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2020] [Accepted: 12/29/2020] [Indexed: 01/23/2023]
Abstract
Huntington's disease (HD) is a progressive neurodegenerative and hyperkinetic movement disorder. Decreased activity of cAMP-responsive element-binding protein (CREB) is thought to contribute to the death of striatal medium spiny neurons in HD. The present study has been designed to explore the possible role of roflumilast against qunilonic acid (QA) induced neurotoxicity in rats intending to investigate whether it inhibits the neuroinflammatory response through activation of the cAMP/CREB/BDNF signaling pathway. QA was microinjected (200 nmol/2 µl, bilaterally) through the intrastriatal route in the stereotaxic apparatus. Roflumilast (0.5, 1, and 2 mg/kg, orally) once-daily treatment for 21 days significantly improved locomotor activity in actophotometer, motor coordination in rotarod, and impaired gait performance in narrow beam walk test. Moreover, roflumilast treatment significantly attenuated oxidative and nitrosative stress (p < 0.05) through attenuating lipid peroxidation nitrite concentration and enhancing reduced glutathione, superoxide dismutase, and catalase levels. Furthermore, roflumilast also significantly decreased elevated pro-inflammatory cytokines like TNF-α (p < 0.01), IL-6 (p < 0.01), IFN-γ (p < 0.05), NF-κB (p < 0.05) and significantly increased BDNF(p < 0.05) in the striatum and cortex of rat brain. The results further demonstrated that roflumilast effectively increased the gene expression of cAMP(p < 0.05), CREB(p < 0.05) and decreased the gene expression of PDE4 (p < 0.05) in qRT-PCR. These results conclusively depicted that roflumilast could be a potential candidate as an effective therapeutic agent in the management of HD through the cAMP/CREB/BDNF signaling pathway.
Collapse
Affiliation(s)
- Priyanka Saroj
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Yashika Bansal
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Raghunath Singh
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Ansab Akhtar
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Rupinder Kaur Sodhi
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India
| | - Mahendra Bishnoi
- National Agri-Food Biotechnology Institue (NABI), Sector 81, SAS Nagar, Mohali, Punjab, 140306, India
| | - Sangeeta Pilkhwal Sah
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India.
- Department of Physiology, University Institute of Pharmaceutical Science (UIPS), UGC Center of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India.
| | - Anurag Kuhad
- Pharmacology Division, University Institute of Pharmaceutical Sciences (UIPS), UGC Centre of Advanced Study (UGC-CAS), Panjab University, Chandigarh, 160014, India.
| |
Collapse
|
9
|
Arora MK, Kisku A, Jangra A. Mangiferin ameliorates intracerebroventricular-quinolinic acid-induced cognitive deficits, oxidative stress, and neuroinflammation in Wistar rats. Indian J Pharmacol 2020; 52:296-305. [PMID: 33078731 PMCID: PMC7722907 DOI: 10.4103/ijp.ijp_699_19] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
INTRODUCTION: Mangiferin (MGF), a xanthonoid polyphenol, confers neuroprotection via combating oxidative stress and inflammation. The current investigation aimed to assess the neuroprotective potential of MGF on behavioral and neurochemical anomalies evoked by administration of quinolinic acid (QA) through intrastriatal injection in male Wistar rats and to reveal the associated mechanisms. MATERIALS AND METHODS: QA (300 nm/4 μl saline) was administered intracerebroventricular in the striatum (unilaterally) once. Thereafter, MGF 20 and 40 mg/kg (peroral) was administered to the animals for 21 days. RESULTS: QA administration caused marked alteration in motor activity (rotatod), footprint analysis, and cognitive function (Morris water maze test, and novel object recognition test). Furthermore, oxido-nitrosative stress (increased nitrite content, lipid peroxidation, with reduction of GSH), cholinergic dysfunction, and mitochondrial complex (I, II, and IV) dysfunction were observed in hippocampus and striatal region of QA-treated rats in comparison to normal control. Pro inflammatory mediators (tumor necrosis factor-alpha TNF-α and interleukin-1β) were noted to increase in the hippocampus and striatum of QA-treated rats. In addition, we observed BDNF depletion in both the hippocampus and striatum of QA-treated animals. MGF treatment significantly ameliorated memory and motor deficits in QA-administered rats. Moreover, MGF treatment (40 mg/kg) restored the GSH level and reduced the MDA, nitrite level, and pro-inflammatory cytokines in striatum and hippocampus. Furthermore, QA-induced cholinergic dysfunction (AChE), BDNF depletion and mitochondrial impairment were found to be ameliorated by MGF treatment. CONCLUSION: The results suggest that MGF offers the neuroprotective potential that may be a promising pharmacological approach to ameliorate cognitive deficits associated with neurodegeneration.
Collapse
Affiliation(s)
- Mandeep Kumar Arora
- Department of Pharmacology, KIET School of Pharmacy, KIET Group of Institutions, Ghaziabad, Uttar Pradesh, India
| | - Anglina Kisku
- Department of Pharmacology, KIET School of Pharmacy, KIET Group of Institutions, Ghaziabad, Uttar Pradesh, India
| | - Ashok Jangra
- Department of Pharmacology, KIET School of Pharmacy, KIET Group of Institutions, Ghaziabad, Uttar Pradesh, India
| |
Collapse
|
10
|
d'Angelo M, Castelli V, Catanesi M, Antonosante A, Dominguez-Benot R, Ippoliti R, Benedetti E, Cimini A. PPARγ and Cognitive Performance. Int J Mol Sci 2019; 20:ijms20205068. [PMID: 31614739 PMCID: PMC6834178 DOI: 10.3390/ijms20205068] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2019] [Revised: 10/09/2019] [Accepted: 10/10/2019] [Indexed: 02/06/2023] Open
Abstract
Recent findings have led to the discovery of many signaling pathways that link nuclear receptors with human conditions, including mental decline and neurodegenerative diseases. PPARγ agonists have been indicated as neuroprotective agents, supporting synaptic plasticity and neurite outgrowth. For these reasons, many PPARγ ligands have been proposed for the improvement of cognitive performance in different pathological conditions. In this review, the research on this issue is extensively discussed.
Collapse
Affiliation(s)
- Michele d'Angelo
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Vanessa Castelli
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Mariano Catanesi
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Andrea Antonosante
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Reyes Dominguez-Benot
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Rodolfo Ippoliti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy
| | - Annamaria Cimini
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy.
- Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University, Philadelphia, PA 19122, USA.
| |
Collapse
|
11
|
Pioglitazone improves working memory performance when administered in chronic TBI. Neurobiol Dis 2019; 132:104611. [PMID: 31513844 DOI: 10.1016/j.nbd.2019.104611] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2019] [Revised: 09/02/2019] [Accepted: 09/06/2019] [Indexed: 01/26/2023] Open
Abstract
Traumatic brain injury (TBI) is a leading cause of long-term disability in the United States. Even in comparatively mild injuries, cognitive and behavioral symptoms can persist for years, and there are currently no established strategies for mitigating symptoms in chronic injury. A key feature of TBI-induced damage in acute and chronic injury is disruption of metabolic pathways. As neurotransmission, and therefore cognition, are highly dependent on the supply of energy, we hypothesized that modulating metabolic activity could help restore behavioral performance even when treatment was initiated weeks after TBI. We treated rats with pioglitazone, a FDA-approved drug for diabetes, beginning 46 days after lateral fluid percussion injury and tested working memory performance in the radial arm maze (RAM) after 14 days of treatment. Pioglitazone treated TBI rats performed significantly better in the RAM test than untreated TBI rats, and similarly to control animals. While hexokinase activity in hippocampus was increased by pioglitazone treatment, there was no upregulation of either the neuronal glucose transporter or hexokinase enzyme expression. Expression of glial markers GFAP and Iba-1 were also not influenced by pioglitazone treatment. These studies suggest that targeting brain metabolism, in particular hippocampal metabolism, may be effective in alleviating cognitive symptoms in chronic TBI.
Collapse
|
12
|
Neuroprotective effect of naringin, a flavone glycoside in quinolinic acid-induced neurotoxicity: Possible role of PPAR-γ, Bax/Bcl-2, and caspase-3. Food Chem Toxicol 2018; 121:95-108. [DOI: 10.1016/j.fct.2018.08.028] [Citation(s) in RCA: 46] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2018] [Revised: 08/12/2018] [Accepted: 08/15/2018] [Indexed: 01/11/2023]
|
13
|
Verma MK, Goel R, Nandakumar K, Nemmani KV. Bilateral quinolinic acid-induced lipid peroxidation, decreased striatal monoamine levels and neurobehavioral deficits are ameliorated by GIP receptor agonist D-Ala 2 GIP in rat model of Huntington's disease. Eur J Pharmacol 2018; 828:31-41. [DOI: 10.1016/j.ejphar.2018.03.034] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2017] [Revised: 02/04/2018] [Accepted: 03/21/2018] [Indexed: 12/19/2022]
|
14
|
L-theanine prevent quinolinic acid induced motor deficit and striatal neurotoxicity: Reduction in oxido-nitrosative stress and restoration of striatal neurotransmitters level. Eur J Pharmacol 2017. [DOI: 10.1016/j.ejphar.2017.06.016] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
|
15
|
Wang H, Zhou J, Liu QZ, Wang LL, Shang J. Simvastatin and Bezafibrate ameliorate Emotional disorder Induced by High fat diet in C57BL/6 mice. Sci Rep 2017; 7:2335. [PMID: 28539670 PMCID: PMC5443827 DOI: 10.1038/s41598-017-02576-5] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2016] [Accepted: 04/13/2017] [Indexed: 12/31/2022] Open
Abstract
High fat diet (HFD)-induced metabolic disorders may lead to emotional disorders. This study aimed to explore the effect of simvastatin (SMV) and bezafibrate (BZ) on improving HFD-induced emotional changes, and tried to identify their different mechanisms. The intraperitoneal glucose tolerance test (IPGTT) was used to evaluate glucose control ability; and behavior tests including open field tests (OFT), forced swimming tests (FST), tail suspension tests (TST) and sucrose preference (SPT), were then performed to evaluate emotional changes. Serum samples were collected for the LC-MS based metabolomics analysis to explore the emotional-related differential compounds; we then evaluated the effect of the drugs. The abnormal serum metabolic profiling and emotional changes caused by HFD in mice was alleviated by SMV treatment, whereas BZ only affected the emotional disorder. The improvement of cannabinoid analogues and then produced influences on the endocannabinoid system, which may be a potential mechanism SMV action. BZ promoted tryptophan-serotonin pathway and inhibited tryptophan-kynurenine pathway, which may be its mechanism of action. Here, we proposed a shed light on the biological mechanisms underlying the observed effects, and identified an important drug candidate for the treatment of emotional disorders induced by HFD.
Collapse
Affiliation(s)
- Hui Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.,Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing, 211198, China
| | - Jia Zhou
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.,Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing, 211198, China
| | - Qiong Zhen Liu
- Qinghai Key Laboratory of Tibetan Medicine Pharmacology and Safety Evaluation, Northwest Institute of Plateau Biology, Chinese Academy of Sciences, Xining, 810008, Qinghai Province, P.R. China
| | - Lu Lu Wang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China.,Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing, 211198, China
| | - Jing Shang
- State Key Laboratory of Natural Medicines, China Pharmaceutical University, Nanjing, 210009, China. .,Jiangsu Key Laboratory of TCM Evaluation and Translational Research, China Pharmaceutical University, Nanjing, 211198, China.
| |
Collapse
|
16
|
Nakatsuji H, Araki A, Hashizume A, Hijikata Y, Yamada S, Inagaki T, Suzuki K, Banno H, Suga N, Okada Y, Ohyama M, Nakagawa T, Kishida K, Funahashi T, Shimomura I, Okano H, Katsuno M, Sobue G. Correlation of insulin resistance and motor function in spinal and bulbar muscular atrophy. J Neurol 2017; 264:839-847. [PMID: 28229243 DOI: 10.1007/s00415-017-8405-3] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2017] [Revised: 01/23/2017] [Accepted: 01/24/2017] [Indexed: 12/22/2022]
Abstract
This study aimed to evaluate various metabolic parameters in patients with spinal and bulbar muscular atrophy (SBMA), to investigate the association between those indices and disease severity, and to explore the underlying molecular pathogenesis. We compared the degree of obesity, metabolic parameters, and blood pressure in 55 genetically confirmed SBMA patients against those in 483 age- and sex-matched healthy control. In SBMA patients, we investigated the correlation between these factors and motor functional indices. SBMA patients had lower body mass index, blood glucose, and Hemoglobin A1c, but higher blood pressure, homeostasis model assessment of insulin resistance (HOMA-IR, a marker of insulin resistance), total cholesterol, and adiponectin levels than the control subjects. There were no differences in visceral fat areas, high-density lipoprotein-cholesterol (HDL-C), or triglyceride levels in two groups. Revised amyotrophic lateral sclerosis functional rating scale (ALSFRS-R) correlated positively with HDL-C, but negatively with HOMA-IR. Through stepwise multiple regression analysis, we identified HOMA-IR as a significant metabolic determinant of ALSFRS-R. In biochemical analysis, we found that decreased expressions of insulin receptors, insulin receptor substrate-1 and insulin receptor-β, in autopsied muscles and fibroblasts of SBMA patients. This study demonstrates that SBMA patients have insulin resistance, which is associated with the disease severity. The expressions of insulin receptors are attenuated in the skeletal muscle of SBMA, providing a possible pathomechanism of metabolic alterations. These findings suggested that insulin resistance is a metabolic index reflecting disease severity and pathogenesis as well as a potential therapeutic target for SBMA.
Collapse
Affiliation(s)
- Hideaki Nakatsuji
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Amane Araki
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.,Department of Neurology, Kasugai Municipal Hospital, Kasugai, Japan
| | - Atsushi Hashizume
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Yasuhiro Hijikata
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Shinichiro Yamada
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Tomonori Inagaki
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Keisuke Suzuki
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.,Department of Clinical Research, Innovation Center for Clinical Research, National Center for Geriatrics and Gerontology, Obu, Japan
| | - Haruhiko Banno
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan
| | - Noriaki Suga
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.,Department of Neurology, Sarashina Rehabilitation Clinic, Ichihara, Japan
| | - Yohei Okada
- Department of Neurology, Aichi Medical University School of Medicine, Aichi, Japan.,Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Manabu Ohyama
- Department of Dermatology, Keio University School of Medicine, Tokyo, Japan
| | - Tohru Nakagawa
- Hitachi, Ltd. Hitachi Health Care Center, Hitachi, Ibaraki, Japan
| | - Ken Kishida
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan.,Kishida Clinic, Osaka, Japan
| | - Tohru Funahashi
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan.,Department of Metabolism and Atherosclerosis, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Iichiro Shimomura
- Department of Metabolic Medicine, Graduate School of Medicine, Osaka University, Osaka, Japan
| | - Hideyuki Okano
- Department of Physiology, Keio University School of Medicine, Tokyo, Japan
| | - Masahisa Katsuno
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| | - Gen Sobue
- Department of Neurology, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan. .,Research Division of Dementia and Neurodegenerative Disease, Nagoya University Graduate School of Medicine, 65 Tsurumai-cho, Showa-ku, Nagoya, 466-8550, Japan.
| |
Collapse
|
17
|
Rosiglitazone activation of PPARγ-dependent signaling is neuroprotective in mutant huntingtin expressing cells. Exp Cell Res 2015; 338:183-93. [DOI: 10.1016/j.yexcr.2015.09.005] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2015] [Revised: 08/26/2015] [Accepted: 09/06/2015] [Indexed: 11/24/2022]
|
18
|
Electron Transport Disturbances and Neurodegeneration: From Albert Szent-Györgyi's Concept (Szeged) till Novel Approaches to Boost Mitochondrial Bioenergetics. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2015; 2015:498401. [PMID: 26301042 PMCID: PMC4537740 DOI: 10.1155/2015/498401] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 03/06/2015] [Accepted: 04/15/2015] [Indexed: 12/21/2022]
Abstract
Impaired function of certain mitochondrial respiratory complexes has long been linked to the pathogenesis of chronic neurodegenerative disorders such as Parkinson's and Huntington's diseases. Furthermore, genetic alterations of mitochondrial genome or nuclear genes encoding proteins playing essential roles in maintaining proper mitochondrial function can lead to the development of severe systemic diseases associated with neurodegeneration and vacuolar myelinopathy. At present, all of these diseases lack effective disease modifying therapy. Following a brief commemoration of Professor Albert Szent-Györgyi, a Nobel Prize laureate who pioneered in the field of cellular respiration, antioxidant processes, and the roles of free radicals in health and disease, the present paper overviews the current knowledge on the involvement of mitochondrial dysfunction in central nervous system diseases associated with neurodegeneration including Parkinson's and Huntington's disease as well as mitochondrial encephalopathies. The review puts special focus on the involvement and the potential therapeutic relevance of peroxisome proliferator-activated receptor-gamma coactivator 1-alpha (PGC-1α), a nuclear-encoded master regulator of mitochondrial biogenesis and antioxidant responses in these disorders, the transcriptional activation of which may hold novel therapeutic value as a more system-based approach aiming to restore mitochondrial functions in neurodegenerative processes.
Collapse
|
19
|
Colín-González AL, Ali SF, Túnez I, Santamaría A. On the antioxidant, neuroprotective and anti-inflammatory properties of S-allyl cysteine: An update. Neurochem Int 2015; 89:83-91. [PMID: 26122973 DOI: 10.1016/j.neuint.2015.06.011] [Citation(s) in RCA: 46] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2015] [Revised: 06/11/2015] [Accepted: 06/15/2015] [Indexed: 12/25/2022]
Abstract
Therapeutic approaches based on isolated compounds obtained from natural products to handle central and peripheral disorders involving oxidative stress and inflammation are more common nowadays. The validation of nutraceutics vs. pharmaceutics as tools to induce preventive and protective profiles in human health alterations is still far of complete acceptance, but the basis to start more solid experimental and clinical protocols with natural products has already begun. S-allyl cysteine (SAC) is a promising garlic-derived organosulfur compound exhibiting a considerable number of positive actions in cell models and living systems. An update, in the form of review, is needed from time to time to get access to the state-of-the-art on this topic. In this review we visited recent and refreshing evidence of new already proven and potential targets to explain the benefits of using SAC against toxic and pathological conditions. The broad spectrum of protective actions covered by this molecule comprises antioxidant, redox modulatory and anti-inflammatory activities, accompanied by anti-apoptotic, pro-energetic and signaling capacities. Herein, we detail the evidence on these aspects to provide the reader a more complete overview on the promising aspects of SAC in research.
Collapse
Affiliation(s)
- Ana Laura Colín-González
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, SSA, Mexico City, Mexico
| | - Syed F Ali
- Neurochemistry Laboratory, Division of Neurotoxicology, National Center for Toxicological Research/FDA, Jefferson, AR, USA.
| | - Isaac Túnez
- Departamento de Bioquímica y Biología Molecular, Facultad de Medicina, Instituto Maimónides de Investigación Biomédica de Córdoba (IMIBIC), Hospital Universitario Reina Sofia/Universidad de Córdoba, Cordoba, Spain; Red Temática de Investigación Cooperativa en Envejecimiento y Fragilidad (RETICEF), Spain
| | - Abel Santamaría
- Laboratorio de Aminoácidos Excitadores, Instituto Nacional de Neurología y Neurocirugía Manuel Velasco Suárez, SSA, Mexico City, Mexico.
| |
Collapse
|
20
|
Huot P, Johnston TH, Fox SH, Brotchie JM. Pioglitazone may impair L-DOPA anti-parkinsonian efficacy in the MPTP-lesioned macaque: Results of a pilot study. Synapse 2015; 69:99-102. [DOI: 10.1002/syn.21801] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2014] [Revised: 12/13/2014] [Accepted: 12/23/2014] [Indexed: 11/11/2022]
Affiliation(s)
- Philippe Huot
- Toronto Western Research Institute, University Health Network; Toronto Ontario Canada
- Division of Neurology, Movement Disorder Clinic; Toronto Western Hospital, University Health Network; Toronto Ontario Canada
| | - Tom H. Johnston
- Toronto Western Research Institute, University Health Network; Toronto Ontario Canada
| | - Susan H. Fox
- Toronto Western Research Institute, University Health Network; Toronto Ontario Canada
- Division of Neurology, Movement Disorder Clinic; Toronto Western Hospital, University Health Network; Toronto Ontario Canada
| | - Jonathan M. Brotchie
- Toronto Western Research Institute, University Health Network; Toronto Ontario Canada
| |
Collapse
|
21
|
Khan A, Jamwal S, Bijjem KRV, Prakash A, Kumar P. Neuroprotective effect of hemeoxygenase-1/glycogen synthase kinase-3β modulators in 3-nitropropionic acid-induced neurotoxicity in rats. Neuroscience 2014; 287:66-77. [PMID: 25536048 DOI: 10.1016/j.neuroscience.2014.12.018] [Citation(s) in RCA: 50] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2014] [Revised: 12/09/2014] [Accepted: 12/11/2014] [Indexed: 01/27/2023]
Abstract
The present study has been designed to explore the possible interaction between hemeoxygenase-1 (HO-1) and glycogen synthase kinase-3β (GSK-3β) pathway in 3-nitropropionic acid (3-NP)-induced neurotoxicity in rats. 3-NP produces neurotoxicity by inhibition of the mitochondrial complex II (enzyme succinate dehydrogenase) and by sensitizing the N-methyl-D-aspartate receptor. Recent studies have reported the therapeutic potential of HO-1/GSK-3β modulators in different neurodegenerative disorders. However, their exact role is yet to be explored. The present study is an attempt to investigate the effect of pharmacological modulation of HO-1/GSK-3β pathway against 3-NP-induced behavioral, biochemical and molecular alterations in rat. Behavioral observation, oxidative stress, pro-inflammatory [tumor necrosis factor-alpha (TNF-α) and interleukin-1 beta (IL-1β)], HO-1 and GSK-3β activity were evaluated post 3-NP treatment. Findings of the present study demonstrate a significant alteration in the locomotor activity, motor coordination, oxidative burden (increased lipid peroxidation, nitrite concentration and decreased endogenous antioxidants), pro-inflammatory mediators [TNF-α, IL-1β], HO-1 and GSK-3β activity in 3-NP-treated animals. Further, administration of hemin (10- and 30-mg/kg; i.p.) and lithium chloride (LiCl) (25- and 50-mg/kg; i.p.) prevented the alteration in body weight, motor impairments, oxidative stress and cellular markers. In addition, combined administration of hemin (10-mg/kg) and LiCl (25-mg/kg) showed synergistic effect on 3-NP-treated rats. Pretreatment with Tin (IV) protoporphyrin (40 μM/kg), HO-1 inhibitor reversed the beneficial effect of LiCl and hemin. Outcomes of the present study suggest that HO-1 and GSK-3β enzymes are involved in the pathophysiology of HD. The modulators of both the pathways might be used as adjuvants or prophylactic therapy for the treatment of HD-like symptoms.
Collapse
Affiliation(s)
- A Khan
- Department of Pharmacology, I.S.F. College of Pharmacy, Ferozepur Road, Ghal Kalan, Moga 142001, Punjab, India
| | - S Jamwal
- Department of Pharmacology, I.S.F. College of Pharmacy, Ferozepur Road, Ghal Kalan, Moga 142001, Punjab, India; Research Scholar, Punjab Technical University, Jalandhar, India
| | - K R V Bijjem
- Department of Pharmacology, I.S.F. College of Pharmacy, Ferozepur Road, Ghal Kalan, Moga 142001, Punjab, India
| | - A Prakash
- Department of Pharmacology, I.S.F. College of Pharmacy, Ferozepur Road, Ghal Kalan, Moga 142001, Punjab, India
| | - P Kumar
- Department of Pharmacology, I.S.F. College of Pharmacy, Ferozepur Road, Ghal Kalan, Moga 142001, Punjab, India.
| |
Collapse
|
22
|
Iida M, Katsuno M, Nakatsuji H, Adachi H, Kondo N, Miyazaki Y, Tohnai G, Ikenaka K, Watanabe H, Yamamoto M, Kishida K, Sobue G. Pioglitazone suppresses neuronal and muscular degeneration caused by polyglutamine-expanded androgen receptors. Hum Mol Genet 2014; 24:314-29. [DOI: 10.1093/hmg/ddu445] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022] Open
|
23
|
Roohafza H, Shokouh P, Sadeghi M, Alikhassy Z, Sarrafzadegan N. A Possible Role for Pioglitazone in the Management of Depressive Symptoms in Metabolic Syndrome Patients (EPICAMP Study): A Double Blind, Randomized Clinical Trial. INTERNATIONAL SCHOLARLY RESEARCH NOTICES 2014; 2014:697617. [PMID: 27433505 PMCID: PMC4897538 DOI: 10.1155/2014/697617] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/13/2014] [Accepted: 06/16/2014] [Indexed: 01/04/2023]
Abstract
The present trial aimed to evaluate the effects of pioglitazone on the mental status of nondiabetic metabolic syndrome patients. From 145 patients screened, 104 eligible volunteers (57% female; age 20-70 years) were enrolled and randomly assigned to receive either pioglitazone (uptitrated to 30 mg/day; P = 53) or matching placebo (P = 51) for 24 weeks. Depression and anxiety were quantified using the hospital anxiety and depression scale and stress level using the general health questionnaire 12 at baseline, week 12, and endpoint. Homeostasis model assessment was used to estimate insulin resistance. At week 24, pioglitazone was superior in mitigating depression score (P = 0.011). In trend analysis, the effect of time (P < 0.001) and group (P = 0.023) as well as the time by group interaction (P = 0.032) on the mean depression score was considerable. In contrast, significant decrements in anxiety and stress levels (P < 0.001 and P = 0.012, resp.) were comparable between two groups. With respect to our findings, alterations in depression severity were not correlated with changes in insulin resistance level (P = 0.145). In conclusion, our findings suggest that pioglitazone might be able to improve mood in nondiabetic insulin resistant patients. (Registered at Australian New Zealand Clinical Trials Registry; ACTRN12611000351910.).
Collapse
Affiliation(s)
- Hamidreza Roohafza
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, P.O. Box 81465-1148, Iran
| | - Pedram Shokouh
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, P.O. Box 81465-1148, Iran
| | - Masoumeh Sadeghi
- Isfahan Cardiac Rehabilitation Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, P.O. Box 81465-1148, Iran
| | - Zahra Alikhassy
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, P.O. Box 81465-1148, Iran
| | - Nizal Sarrafzadegan
- Isfahan Cardiovascular Research Center, Cardiovascular Research Institute, Isfahan University of Medical Sciences, Isfahan, P.O. Box 81465-1148, Iran
| |
Collapse
|
24
|
Aghaei I, Shabani M, Doustar N, Nazeri M, Dehpour A. Peroxisome proliferator-activated receptor-γ activation attenuates motor and cognition impairments induced by bile duct ligation in a rat model of hepatic cirrhosis. Pharmacol Biochem Behav 2014; 120:133-9. [DOI: 10.1016/j.pbb.2014.03.002] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/10/2014] [Revised: 02/14/2014] [Accepted: 03/01/2014] [Indexed: 12/21/2022]
|
25
|
Prakash A, Kumar A. Pioglitazone alleviates the mitochondrial apoptotic pathway and mito-oxidative damage in the d-galactose-induced mouse model. Clin Exp Pharmacol Physiol 2014; 40:644-51. [PMID: 23795773 DOI: 10.1111/1440-1681.12144] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/23/2012] [Revised: 06/16/2013] [Accepted: 06/18/2013] [Indexed: 11/25/2022]
Abstract
Chronic injection of d-galactose can cause gradual deterioration in learning and memory capacity, and activates oxidative stress, mitochondrial dysfunction and apoptotic cell death in the brain of mice. Thus, it serves as an animal model of ageing. Recent evidence has shown that mild cognitive impairment in humans might be alleviated by treatment with piogliatzone (peroxisome proliferator-activated receptor gamma (PPARγ) agonists). To continue exploring the effects of piogliatzone in this model, we focused on behavioural alteration, oxidative damage, mitochondrial dysfunction and apoptosis in d-galactose-induced mice. The ageing model was established by administration of d-galactose (100 mg/kg) for 6 weeks. Pioglitazone (10 and 30 mg/kg) and bisphenol A diglycidyl ether (15 mg/kg) were given daily to d-galactose-induced senescent mice. The cognitive behaviour of mice was monitored using the Morris water maze. The anti-oxidant status and apoptotic activity in the ageing mice was measured by determining mito-oxidative parameters and caspase-3 activity in brain tissue. Systemic administration of d-galactose significantly increased behavioural alterations, biochemical parameters, mitochondrial enzymes, and activations of caspase-3 and acetylcholinesterase enzyme activity as compared with the control group. Piogliatzone treatment significantly improved behavioural abnormalities, biochemical, cellular alterations, and attenuated the caspase-3 and acetylcholinesterase enzyme activity as compared with the control. Furthermore, pretreatment of BADGE (PPARγ antagonist) with pioglitazone reversed the protective effect of pioglitazone in d-galactose-induced mice. The present study highlights the protective effects of pioglitzone against d-galactose-induced memory dysfunction, mito-oxidative damage and apoptosis through activation of PPARγ receptors. These findings suggest that pioglitazone might be helpful for the prevention or alleviation of ageing.
Collapse
Affiliation(s)
- Atish Prakash
- Mental Health Research Institute, The University of Melbourne, Melbourne, VIC, Australia.
| | | |
Collapse
|
26
|
Hsiao HY, Chiu FL, Chen CM, Wu YR, Chen HM, Chen YC, Kuo HC, Chern Y. Inhibition of soluble tumor necrosis factor is therapeutic in Huntington's disease. Hum Mol Genet 2014; 23:4328-44. [PMID: 24698979 DOI: 10.1093/hmg/ddu151] [Citation(s) in RCA: 79] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Neuroinflammation is a common feature of many neurodegenerative diseases, including Huntington's disease (HD). HD is an autosomal dominant genetic disease caused by an expanded CAG repeat in exon 1 of the huntingtin (HTT) gene. Previous studies demonstrated that levels of several proinflammatory cytokines, including tumor necrosis factor (TNF)-α, were higher in the plasma and brain tissues of mice and patients with HD, suggesting that inflammation may contribute to HD progression. To evaluate the pathological role of TNF-α in HD pathogenesis, we blocked TNF-α signaling using a dominant negative inhibitor of soluble TNF-α (XPro1595). XPro1595 effectively suppressed the inflammatory responses of primary astrocytes-enriched culture isolated from a transgenic mouse model (R6/2) and human astrocytes-enriched culture derived from induced pluripotent stem cells (iPSCs) of HD patients evoked by lipopolysaccharide and cytokines, respectively. Moreover, XPro1595 protected the cytokine-induced toxicity of primary R6/2 neurons and human neurons derived from iPSCs of HD patients. To assess the beneficial effect of XPro1595 in vivo, an intracerebroventricular (i.c.v.) infusion was provided with an osmotic minipump. ELISA analyses showed that i.c.v. infusion of XPro1595 decreased elevated levels of TNFα in the cortex and striatum, improved motor function, reduced caspase activation, diminished the amount of mutant HTT aggregates, increased neuronal density and decreased gliosis in brains of R6/2 mice. Moreover, reducing the peripheral inflammatory response by a systemic injection of XPro1595 improved the impaired motor function of R6/2 mice but did not affect caspase activation. Collectively, our findings suggest that an effective and selective anti-inflammatory treatment targeting the abnormal brain inflammatory response is a potential therapeutic strategy for HD.
Collapse
Affiliation(s)
- Han-Yun Hsiao
- Institute of Biomedical Sciences Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan
| | | | - Chiung-Mei Chen
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center and College of Medicine, Chang-Gung University, Taoyuan, Taiwan
| | - Yih-Ru Wu
- Department of Neurology, Chang Gung Memorial Hospital, Linkou Medical Center and College of Medicine, Chang-Gung University, Taoyuan, Taiwan
| | | | - Yu-Chen Chen
- Institute of Biomedical Sciences Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan
| | - Hung-Chih Kuo
- Institute of Cellular and Organismic Biology and Genomics Research Center, Academia Sinica, Taipei 115, Taiwan
| | - Yijuang Chern
- Institute of Biomedical Sciences Institute of Neuroscience, National Yang-Ming University, Taipei, Taiwan
| |
Collapse
|
27
|
Mishra J, Chaudhary T, Kumar A. Rosiglitazone synergizes the neuroprotective effects of valproic acid against quinolinic acid-induced neurotoxicity in rats: targeting PPARγ and HDAC pathways. Neurotox Res 2014; 26:130-51. [PMID: 24566814 DOI: 10.1007/s12640-014-9458-z] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2013] [Revised: 02/01/2014] [Accepted: 02/04/2014] [Indexed: 12/15/2022]
Abstract
Huntington's disease (HD) is an autosomal dominant progressive neurodegenerative disorder which affects medium spiny GABAergic neurons mainly in the striatum. Oxidative damage, neuro-inflammation, apoptosis, protein aggregation, and signaling of neurotrophic factors are some of the common cellular pathways involved in HD. Quinolinic acid (QA) causes excitotoxicity by stimulating N-methyl-D-aspartate receptors via calcium overload leading to neurodegeneration. Neuroprotective potential of peroxisome proliferator activated receptor-γ (PPARγ) agonists and histone deacetylase (HDAC) inhibitors have been well documented in experimental models of neurodegenerative disorders; however, their exact mechanisms are not clear. Therefore, present study has been designed to explore possible neuroprotective mechanism of valproic acid (VPA) and its interaction with rosiglitazone against QA induced HD-like symptoms in rats. Single bilateral intrastriatal QA (200 nmol/2 μl saline) administration significantly caused motor incoordination, memory impairment, oxidative damage, mitochondrial dysfunction (complex I, II, II and IV), cellular alterations [tumor necrosis factor-alpha (TNF-α), caspase-3, brain derived neurotrophic factor, acetylcholinesterase], and striatal neurodegeneration as compared to sham group. Treatment with rosiglitazone (5, 10 mg/kg) and VPA (100, 200 mg/kg) for 21 days significantly attenuated these behavioral, biochemical, and cellular alterations as compared to control (QA 200 nmol) group. However, VPA (100 mg/kg) treatment in combination with rosiglitazone (5 mg/kg) for 21 days synergized their neuroprotective effect, which was significant as compared to their effects per se in QA-treated animals. The present study provides an evidence of possible interplay of PPARγ agonists and HDAC inhibitors as a novel therapeutic strategy in the management of HD.
Collapse
Affiliation(s)
- Jitendriya Mishra
- Pharmacology Division, UGC Centre of Advanced Study (UGC-CAS), University Institute of Pharmaceutical Sciences, Panjab University, Chandigarh, 160014, India
| | | | | |
Collapse
|
28
|
Role of neurosteroids in experimental 3-nitropropionic acid induced neurotoxicity in rats. Eur J Pharmacol 2013; 723:38-45. [PMID: 24333475 DOI: 10.1016/j.ejphar.2013.11.036] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2013] [Revised: 11/27/2013] [Accepted: 11/28/2013] [Indexed: 11/22/2022]
Abstract
Huntington's disease is an autosomal dominant, progressive, and fatal neurodegenerative disease characterized by motor and non-motor symptoms. Systemic administration of 3-nitropropionic acid, a complex II inhibitor of the electron transport chain induces selective striatal lesions in rodents. Neurosteroids are synthesized in central nervous system, able to modulate GABAA receptor function and has been reported to have neuroprotective action. The present study has been designed to investigate the role of neurosteroids such as progesterone and pregnenolone which are positive and negative modulators of GABA respectively against 3-nitropropionic acid induced experimental Huntington's disease. Systemic administration of 3-nitropropionic acid (10mg/kg i.p.) for 14 days significantly reduced body weight, locomotor activity, motor coordination, balance beam walk performance, antioxidant defense enzymes (reduced glutathione and catalase) and significantly increase oxidative stress markers (lipid peroxidation and nitrite level) in striatum and cortex. 3-Nitropropionic acid treatment also increases pro-inflammatory cytokines (TNF-α and IL-1β) level in striatum. Progesterone (10, 20mg/kg/day i.p.) treatments for 14 days significantly reversed the behavioral, antioxidant defense enzymes, oxidative stress marker and pro-inflammatory cytokines as compared to the 3-Nitropropionic acid treated group. Pregnenolone (1 and 2mg/kg i.p.), a negative modulator of GABAA pretreatment significantly reversed the protective effect of progesterone on behavioral and biochemical parameters. The results of the present study suggest that the positive GABAergic modulation may be beneficial for the treatment of motor disorder.
Collapse
|
29
|
Grover S, Kumar P, Singh K, Vikram V, Budhiraja R. Possible beneficial effect of peroxisome proliferator-activated receptor (PPAR) — α and γ agonist against a rat model of oral dyskinesia. Pharmacol Biochem Behav 2013; 111:17-23. [DOI: 10.1016/j.pbb.2013.08.001] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Revised: 07/26/2013] [Accepted: 08/01/2013] [Indexed: 10/26/2022]
|
30
|
Quinolinic acid: an endogenous neurotoxin with multiple targets. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2013; 2013:104024. [PMID: 24089628 PMCID: PMC3780648 DOI: 10.1155/2013/104024] [Citation(s) in RCA: 417] [Impact Index Per Article: 34.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/14/2013] [Revised: 07/23/2013] [Accepted: 08/01/2013] [Indexed: 11/21/2022]
Abstract
Quinolinic acid (QUIN), a neuroactive metabolite of the kynurenine pathway, is normally presented in nanomolar concentrations in human brain and cerebrospinal fluid (CSF) and is often implicated in the pathogenesis of a variety of human neurological diseases. QUIN is an agonist of N-methyl-D-aspartate (NMDA) receptor, and it has a high in vivo potency as an excitotoxin. In fact, although QUIN has an uptake system, its neuronal degradation enzyme is rapidly saturated, and the rest of extracellular QUIN can continue stimulating the NMDA receptor. However, its toxicity cannot be fully explained by its activation of NMDA receptors it is likely that additional mechanisms may also be involved. In this review we describe some of the most relevant targets of QUIN neurotoxicity which involves presynaptic receptors, energetic dysfunction, oxidative stress, transcription factors, cytoskeletal disruption, behavior alterations, and cell death.
Collapse
|
31
|
Kashani L, Omidvar T, Farazmand B, Modabbernia A, Ramzanzadeh F, Tehraninejad ES, Ashrafi M, Tabrizi M, Akhondzadeh S. Does pioglitazone improve depression through insulin-sensitization? Results of a randomized double-blind metformin-controlled trial in patients with polycystic ovarian syndrome and comorbid depression. Psychoneuroendocrinology 2013; 38:767-76. [PMID: 22999261 DOI: 10.1016/j.psyneuen.2012.08.010] [Citation(s) in RCA: 88] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/17/2012] [Revised: 08/24/2012] [Accepted: 08/28/2012] [Indexed: 12/28/2022]
Abstract
Thiazolidinediones have shown beneficial effects in short-term treatment of depression. However, it is unclear whether the antidepressant efficacy of these agents is related to their insulin-sensitizing action. We conducted the present study to compare the antidepressant efficacy of pioglitazone with another insulin-sensitizer, metformin, in obese patients with concomitant polycystic ovarian syndrome (PCOS) and major depressive disorder (MDD). In a six-week double-blind study, 50 patients with PCOS and MDD (DSM-IV-TR criteria) with Hamilton depression rating scale (HDRS) score of <20, randomly received pioglitazone (15 mg twice daily; PO) or metformin (750 mg twice daily; PO). Assessment was done using HDRS (weeks 0, 3, 6) together with fasting Insulin, glucose, and lipid profile, liver enzymes, homeostatic model assessment of insulin resistance (HOMA-IR), anthropometric measures, and serum androgens (weeks 0 and 6). Pioglitazone was superior to metformin in reducing HDRS scores at the end of the study [38.3% versus 8.3% reduction from baseline scores, F(1, 37) = 73.513, P<0.001]. Changes from baseline in HOMA-IR values at week 6 were not significantly different between the two groups (P = 0.888). Baseline (but not follow-up) HDRS and HOMA-IR values were significantly correlated (r = 0.393, P = 0.012). In multiple regression analysis, treatment with pioglitazone independent of HOMA-IR values predicted greater score reduction on HDRS at week 6 (standardized beta = 0.801, P<0.001). Biochemical and hormonal profile did not differ between the two groups at week 6. Metformin was associated with higher frequency of gastrointestinal side effects (P = 0.014). In summary, we showed that pioglitazone improved depression with mechanisms largely unrelated to its insulin-sensitizing action (registration number: IRCT201106081556N23).
Collapse
Affiliation(s)
- Ladan Kashani
- Infertility Ward, Arash Hospital, Tehran University of Medical Sciences, Tehran, Iran
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Chen GL, Miller GM. Extensive alternative splicing of the repressor element silencing transcription factor linked to cancer. PLoS One 2013; 8:e62217. [PMID: 23614038 PMCID: PMC3628349 DOI: 10.1371/journal.pone.0062217] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2013] [Accepted: 03/18/2013] [Indexed: 12/15/2022] Open
Abstract
The repressor element silencing transcription factor (REST) is a coordinate transcriptional and epigenetic regulator which functions as a tumor suppressor or an oncogene depending on cellular context, and a truncated splice variant REST4 has been linked to various types of cancer. We performed a comprehensive analysis of alternative splicing (AS) of REST by rapid amplification of cDNA ends and PCR amplification of cDNAs from various tissues and cell lines with specific primers. We identified 8 novel alternative exons including an alternate last exon which doubles the REST gene boundary, along with numerous 5'/3' splice sites and ends in the constitutive exons. With the combination of various splicing patterns (e.g. exon skipping and alternative usage of the first and last exons) that are predictive of altered REST activity, at least 45 alternatively spliced variants of coding and non-coding mRNA were expressed in a species- and cell-type/tissue-specific manner with individual differences. By examining the repertoire of REST pre-mRNA splicing in 27 patients with kidney, liver and lung cancer, we found that all patients without exception showed differential expression of various REST splice variants between paired tumor and adjacent normal tissues, with striking cell-type/tissue and individual differences. Moreover, we revealed that exon 3 skipping, which causes no frame shift but loss of a domain essential for nuclear translocation, was affected by pioglitazone, a highly selective activator of the peroxisome proliferator-activated receptor gamma (PPARγ) which contributes to cell differentiation and tumorigenesis besides its metabolic actions. Accordingly, this study demonstrates an extensive AS of REST pre-mRNA which redefines REST gene boundary and structure, along with a general but differential link between REST pre-mRNA splicing and various types of cancer. These findings advance our understanding of the complex, context-dependent regulation of REST gene expression and function, and provide potential biomarkers and therapeutic targets for cancer.
Collapse
Affiliation(s)
- Guo-Lin Chen
- Division of Neuroscience, New England Primate Research Center, Harvard Medical School, Southborough, Massachusetts, United States of America.
| | | |
Collapse
|
33
|
Yin QQ, Pei JJ, Xu S, Luo DZ, Dong SQ, Sun MH, You L, Sun ZJ, Liu XP. Pioglitazone improves cognitive function via increasing insulin sensitivity and strengthening antioxidant defense system in fructose-drinking insulin resistance rats. PLoS One 2013; 8:e59313. [PMID: 23527159 PMCID: PMC3603906 DOI: 10.1371/journal.pone.0059313] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2012] [Accepted: 02/13/2013] [Indexed: 12/18/2022] Open
Abstract
Insulin resistance (IR) links Alzheimer’s disease (AD) with oxidative damage, cholinergic deficit, and cognitive impairment. Peroxisome proliferator-activated receptor γ (PPARγ) agonist pioglitazone previously used to treat type 2 diabetes mellitus (T2DM) has also been demonstrated to be effective in anti-inflammatory reaction and anti-oxidative stress in the animal models of AD and other neuroinflammatory diseases. Here, we investigated the effect of pioglitazone on learning and memory impairment and the molecular events that may cause it in fructose-drinking insulin resistance rats. We found that long-term fructose-drinking causes insulin resistance, oxidative stress, down-regulated activity of cholinergic system, and cognitive deficit, which could be ameliorated by pioglitazone administration. The results from the present study provide experimental evidence for using pioglitazone in the treatment of brain damage caused by insulin resistance.
Collapse
Affiliation(s)
- Qing-Qing Yin
- Department of Senile Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
| | - Jin-Jing Pei
- Department of KI-Alzheimer Disease Research Center, Karolinska Institutet, Stockholm, Sweden
| | - Song Xu
- Department of Anti-Ageing, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
| | - Ding-Zhen Luo
- Department of Senile Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
| | - Si-Qing Dong
- Department of Senile Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
| | - Meng-Han Sun
- Department of Senile Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
| | - Li You
- Department of Central Lab, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
| | - Zhi-Jian Sun
- Department of Anti-Ageing, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
| | - Xue-Ping Liu
- Department of Senile Neurology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
- Department of Anti-Ageing, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong, P. R. China
- * E-mail:
| |
Collapse
|
34
|
Dumont M, Stack C, Elipenahli C, Jainuddin S, Gerges M, Starkova N, Calingasan NY, Yang L, Tampellini D, Starkov AA, Chan RB, Di Paolo G, Pujol A, Beal MF. Bezafibrate administration improves behavioral deficits and tau pathology in P301S mice. Hum Mol Genet 2012; 21:5091-105. [PMID: 22922230 PMCID: PMC3490516 DOI: 10.1093/hmg/dds355] [Citation(s) in RCA: 58] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/27/2012] [Accepted: 08/17/2012] [Indexed: 12/11/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) are ligand-mediated transcription factors, which control both lipid and energy metabolism and inflammation pathways. PPARγ agonists are effective in the treatment of metabolic diseases and, more recently, neurodegenerative diseases, in which they show promising neuroprotective effects. We studied the effects of the pan-PPAR agonist bezafibrate on tau pathology, inflammation, lipid metabolism and behavior in transgenic mice with the P301S human tau mutation, which causes familial frontotemporal lobar degeneration. Bezafibrate treatment significantly decreased tau hyperphosphorylation using AT8 staining and the number of MC1-positive neurons. Bezafibrate treatment also diminished microglial activation and expression of both inducible nitric oxide synthase and cyclooxygenase 2. Additionally, the drug differentially affected the brain and brown fat lipidome of control and P301S mice, preventing lipid vacuoles in brown fat. These effects were associated with behavioral improvement, as evidenced by reduced hyperactivity and disinhibition in the P301S mice. Bezafibrate therefore exerts neuroprotective effects in a mouse model of tauopathy, as shown by decreased tau pathology and behavioral improvement. Since bezafibrate was given to the mice before tau pathology had developed, our data suggest that bezafibrate exerts a preventive effect on both tau pathology and its behavioral consequences. Bezafibrate is therefore a promising agent for the treatment of neurodegenerative diseases associated with tau pathology.
Collapse
Affiliation(s)
- Magali Dumont
- Department of Neurology and Neuroscience, Weill Cornell Medical College, New York, NY 10065, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Katsouri L, Blondrath K, Sastre M. Peroxisome proliferator-activated receptor-γ cofactors in neurodegeneration. IUBMB Life 2012; 64:958-64. [DOI: 10.1002/iub.1097] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/13/2012] [Accepted: 09/19/2012] [Indexed: 12/21/2022]
|
36
|
Kemp DE, Ismail-Beigi F, Ganocy SJ, Conroy C, Gao K, Obral S, Fein E, Findling RL, Calabrese JR. Use of insulin sensitizers for the treatment of major depressive disorder: a pilot study of pioglitazone for major depression accompanied by abdominal obesity. J Affect Disord 2012; 136:1164-73. [PMID: 21782251 PMCID: PMC3225727 DOI: 10.1016/j.jad.2011.06.033] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/14/2011] [Revised: 06/07/2011] [Accepted: 06/23/2011] [Indexed: 12/21/2022]
Abstract
OBJECTIVE This study was conducted to examine the safety and efficacy of pioglitazone, a thiazolidinedione insulin sensitizer, in adult outpatients with major depressive disorder. METHOD In a 12-week, open-label, flexible-dose study, 23 patients with major depressive disorder received pioglitazone monotherapy or adjunctive therapy initiated at 15 mg daily. Subjects were required to meet criteria for abdominal obesity (waist circumference>35 in. in women and >40 in. in men) or metabolic syndrome. The primary efficacy measure was the change from baseline to Week 12 on the Inventory of Depressive Symptomatology (IDS) total score. Partial responders (≥25% decrease in IDS total score) were eligible to participate in an optional extension phase for an additional three months. RESULTS Pioglitazone decreased depression symptom severity from a total IDS score of 40.3±1.8 to 19.2±1.8 at Week 12 (p<.001). Among partial responders (≥25% decrease in IDS total score), an improvement in depressive symptoms was maintained during an additional 3-month extension phase (total duration=24 weeks) according to IDS total scores (p<.001). Patients experienced a reduction in insulin resistance from baseline to Week 12 according to the log homeostasis model assessment (-0.8±0.75; p<.001) and a significant reduction in inflammation as measured by log highly- sensitive C-reactive protein (-0.87±0.72; p<.001). During the current episode, the majority of participants (74%, n=17), had already failed at least one antidepressant trial. The most common side effects were headache and dizziness; no patient discontinued due to side effects. LIMITATIONS These data are limited by a small sample size and an open-label study design with no placebo control. CONCLUSION Although preliminary, pioglitazone appears to reduce depression severity and improve several markers of cardiometabolic risk, including insulin resistance and inflammation. Larger, placebo-controlled studies are indicated.
Collapse
Affiliation(s)
- David E. Kemp
- Case Western Reserve University, University Hospitals Case Medical Center, Department of Psychiatry, Cleveland, OH, USA
| | - Faramarz Ismail-Beigi
- University Hospitals Case Medical Center and Cleveland VA Medical Center, Cleveland, OH, USA
| | - Stephen J. Ganocy
- Case Western Reserve University, University Hospitals Case Medical Center, Department of Psychiatry, Cleveland, OH, USA
| | - Carla Conroy
- Case Western Reserve University, University Hospitals Case Medical Center, Department of Psychiatry, Cleveland, OH, USA
| | - Keming Gao
- Case Western Reserve University, University Hospitals Case Medical Center, Department of Psychiatry, Cleveland, OH, USA
| | - Sarah Obral
- Case Western Reserve University, University Hospitals Case Medical Center, Department of Psychiatry, Cleveland, OH, USA
| | - Elizabeth Fein
- Case Western Reserve University, University Hospitals Case Medical Center, Department of Psychiatry, Cleveland, OH, USA
| | - Robert L. Findling
- Case Western Reserve University, University Hospitals Case Medical Center, Department of Psychiatry, Cleveland, OH, USA
| | - Joseph R. Calabrese
- Case Western Reserve University, University Hospitals Case Medical Center, Department of Psychiatry, Cleveland, OH, USA
| |
Collapse
|
37
|
Kumar P, Kalonia H, Kumar A. Possible GABAergic mechanism in the neuroprotective effect of gabapentin and lamotrigine against 3-nitropropionic acid induced neurotoxicity. Eur J Pharmacol 2012; 674:265-74. [DOI: 10.1016/j.ejphar.2011.11.030] [Citation(s) in RCA: 56] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2011] [Revised: 11/12/2011] [Accepted: 11/16/2011] [Indexed: 11/29/2022]
|
38
|
Johri A, Beal MF. Antioxidants in Huntington's disease. Biochim Biophys Acta Mol Basis Dis 2011; 1822:664-74. [PMID: 22138129 DOI: 10.1016/j.bbadis.2011.11.014] [Citation(s) in RCA: 110] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/12/2011] [Revised: 11/11/2011] [Accepted: 11/12/2011] [Indexed: 02/08/2023]
Abstract
Huntington's disease (HD) is a prototypical neurodegenerative disease in which there is selective neuronal degeneration, which leads to progressive disability, manifesting itself as a movement disorder, with both psychiatric and cognitive impairment. The disease is caused by a cytosine-adenine-guanine (CAG) repeat expansion in the huntingtin gene, which causes an expanded polyglutamine repeat in the huntingtin protein, resulting in a protein with a novel gain of function. The mutant huntingtin protein causes neuronal dysfunction and eventual cell death in which transcriptional impairment, excitotoxicity, oxidative damage, inflammation, apoptosis and mitochondrial dysfunction are all implicated. A critical transcriptional impairment may be impaired expression and function of peroxisome proliferator-activated receptor gamma coactivator-1α (PGC-1α), a master co-regulator of mitochondrial biogenesis and expression of antioxidant enzymes. A deficiency of PGC-1α leads to increased vulnerability to oxidative stress and to striatal degeneration. The extent and severity of the oxidative damage in HD are features well recognized but perhaps under-appreciated. Oxidative damage occurs to lipids, proteins and deoxyribonucleic acid (DNA), and it has been suggested that the latter may contribute to CAG repeat expansion during DNA repair [1]. A marked elevation of oxidized DNA bases occurs in patients' plasma, which may provide a biomarker of disease progression. Antioxidants are effective in slowing disease progression in transgenic mouse models of HD, and show promise in human clinical trials. Strategies to transcriptionally increase expression of antioxidant enzymes by modulating the Nrf-2/ARE pathway, or by increasing expression of PGC-1α hold great promise for developing new treatments to slow or halt the progression of HD. This article is part of a Special Issue entitled: Antioxidants and Antioxidant Treatment in Disease.
Collapse
Affiliation(s)
- Ashu Johri
- Department of Neurology and Neuroscience, Weill Medical College of Cornell University, New York-Presbyterian Hospital, New York, NY 10065, USA.
| | | |
Collapse
|
39
|
Signaling mechanisms downstream of quinolinic acid targeting the cytoskeleton of rat striatal neurons and astrocytes. Exp Neurol 2011; 233:391-9. [PMID: 22116044 DOI: 10.1016/j.expneurol.2011.11.005] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2011] [Revised: 10/31/2011] [Accepted: 11/07/2011] [Indexed: 11/22/2022]
Abstract
The studies of signaling mechanisms involved in the disruption of the cytoskeleton homeostasis were performed in a model of quinolinic acid (QUIN) neurotoxicity in vitro. This investigation focused on the phosphorylation level of intermediate filament (IF) subunits of astrocytes (glial fibrillary acidic protein - GFAP) and neurons (low, medium and high molecular weight neurofilament subunits - NFL, NFM and NFH, respectively). The activity of the phosphorylating system associated with the IFs was investigated in striatal slices of rat exposed to QUIN or treated simultaneously with QUIN plus glutamate receptor antagonists, calcium channel blockers or kinase inhibitors. Results showed that in astrocytes, the action of 100 μM QUIN was mainly due to increased Ca(2+) influx through NMDA and L-type voltage-dependent Ca(2+) channels (L-VDCC). In neuronal cells QUIN acted through metabotropic glutamate receptor (mGluR) activation and influx of Ca(2+) through NMDA receptors and L-VDCC, as well as Ca(2+) release from intracellular stores. These mechanisms then set off a cascade of events including activation of PKA, PKCaMII and PKC, which phosphorylate head domain sites on GFAP and NFL. Also, Cdk5 was activated downstream of mGluR5, phosphorylating the KSP repeats on NFM and NFH. mGluR1 was upstream of phospholipase C (PLC) which, in turn, produced diacylglycerol (DAG) and inositol 3,4,5 triphosphate (IP3). DAG is important to activate PKC and phosphorylate NFL, while IP(3) contributed to Ca(2+) release from internal stores promoting hyperphosphorylation of KSP repeats on the tail domain of NFM and NFH. The present study supports the concept of glutamate and Ca(2+) contribution in excitotoxic neuronal damage provoked by QUIN associated to dysfunction of the cytoskeleton homeostasis and highlights the differential signaling mechanisms elicited in striatal astrocytes and neurons.
Collapse
|
40
|
Pharmacological preconditioning with nicorandil and pioglitazone attenuates myocardial ischemia/reperfusion injury in rats. Eur J Pharmacol 2011; 663:51-8. [PMID: 21549700 DOI: 10.1016/j.ejphar.2011.04.038] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2010] [Revised: 02/22/2011] [Accepted: 04/14/2011] [Indexed: 12/18/2022]
Abstract
The present investigation was designed to study the cardioprotective effects of nicorandil and pioglitazone preconditioning in myocardial ischemia/reperfusion-induced hemodynamic, biochemical and histological changes in rats. Oral doses of nicorandil (3 or 6 mg/kg) and pioglitazone (10 or 20mg/kg) were administered once daily for 5 consecutive days. Rats were then subjected to myocardial ischemia/reperfusion (40 min/10 min). Heart rate and ventricular arrhythmias were recorded during ischemia/reperfusion progress. At the end of reperfusion, plasma creatine kinase-MB activity and total nitrate/nitrite were determined. In addition, lactate, adenine nucleotides, thiobarbituric acid reactive substances, reduced glutathione and myeloperoxidase activity were estimated in the heart left ventricle. Finally, histological examination was performed to visualize the protective cellular effects of different pretreatments. Nicorandil (3 or 6 mg/kg) was effective in attenuating the ischemia/reperfusion-induced ventricular arrhythmias, creatine kinase-MB release, lactate accumulation and oxidative stress. Nicorandil (3 mg/kg) was more effective in improving the energy production and lowering the elevated myeloperoxidase activity. Both doses of pioglitazone (10 or 20 mg/kg) were equally effective in reducing lactate accumulation and completely counteracting the oxidative stress. Pioglitazone (10 mg/kg) was more effective in improving energy production and reducing ventricular arrhythmias, plasma creatine kinase-MB release and total nitrate/nitrite. It seems that selective mitochondrial K(ATP) channel opening by lower doses of nicorandil and pioglitazone in the present study provided more cardioprotection against ventricular arrhythmias and biochemical changes induced by ischemia/reperfusion. Histological examination revealed also better improvement by the lower dose of nicorandil than that of pioglitazone.
Collapse
|
41
|
Licofelone attenuates quinolinic acid induced Huntington like symptoms: possible behavioral, biochemical and cellular alterations. Prog Neuropsychopharmacol Biol Psychiatry 2011; 35:607-15. [PMID: 21237233 DOI: 10.1016/j.pnpbp.2011.01.003] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2010] [Revised: 12/08/2010] [Accepted: 01/05/2011] [Indexed: 11/22/2022]
Abstract
Cyclo-oxygenase and lipoxygenase enzymes are involved in arachidonic acid metabolism. Emerging evidence indicates that cyclo-oxygenase and lipoxygenase inhibitors prevent neurodegenerative processes and related complications. Therefore, the present study has been designed to explore the neuroprotective potential of licofelone (dual COX-2/5-LOX inhibitor) against quinolinic acid induced Huntington like symptom in rats. Intrastriatal administration of quinolinic acid significantly caused reduction in body weight and motor function (locomotor activity, rotarod performance and beam walk test), oxidative defense (as evidenced by increased lipid peroxidation, nitrite concentration and decreased endogenous antioxidant enzymes), alteration in mitochondrial enzyme complex (I, II and IV) activities, raised TNF-α level and striatal lesion volume as compared to sham treated animals. Licofelone (2.5, 5 and 10 mg/kg) treatment significantly improved body weight, locomotor activity, rotarod performance, balance beam walk performance, oxidative defense, mitochondrial enzyme complex activities and attenuated TNF-α level and striatal lesion as compared to control (quinolinic acid). The present study highlights that licofelone attenuates behavioral, biochemical and cellular alterations against quinolinic acid induced neurotoxicity and this could be an important therapeutic avenue to ameliorate the Huntington like symptoms.
Collapse
|