101
|
Balakrishnan R, Azam S, Cho DY, Su-Kim I, Choi DK. Natural Phytochemicals as Novel Therapeutic Strategies to Prevent and Treat Parkinson's Disease: Current Knowledge and Future Perspectives. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2021; 2021:6680935. [PMID: 34122727 PMCID: PMC8169248 DOI: 10.1155/2021/6680935] [Citation(s) in RCA: 33] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/16/2020] [Revised: 04/14/2021] [Accepted: 04/26/2021] [Indexed: 12/12/2022]
Abstract
Parkinson's disease (PD) is the second-most common neurodegenerative chronic disease affecting both cognitive performance and motor functions in aged people. Yet despite the prevalence of this disease, the current therapeutic options for the management of PD can only alleviate motor symptoms. Research has explored novel substances for naturally derived antioxidant phytochemicals with potential therapeutic benefits for PD patients through their neuroprotective mechanism, targeting oxidative stress, neuroinflammation, abnormal protein accumulation, mitochondrial dysfunction, endoplasmic reticulum stress, neurotrophic factor deficit, and apoptosis. The aim of the present study is to perform a comprehensive evaluation of naturally derived antioxidant phytochemicals with neuroprotective or therapeutic activities in PD, focusing on their neuropharmacological mechanisms, including modulation of antioxidant and anti-inflammatory activity, growth factor induction, neurotransmitter activity, direct regulation of mitochondrial apoptotic machinery, prevention of protein aggregation via modulation of protein folding, modification of cell signaling pathways, enhanced systemic immunity, autophagy, and proteasome activity. In addition, we provide data showing the relationship between nuclear factor E2-related factor 2 (Nrf2) and PD is supported by studies demonstrating that antiparkinsonian phytochemicals can activate the Nrf2/antioxidant response element (ARE) signaling pathway and Nrf2-dependent protein expression, preventing cellular oxidative damage and PD. Furthermore, we explore several experimental models that evaluated the potential neuroprotective efficacy of antioxidant phytochemical derivatives for their inhibitory effects on oxidative stress and neuroinflammation in the brain. Finally, we highlight recent developments in the nanodelivery of antioxidant phytochemicals and its neuroprotective application against pathological conditions associated with oxidative stress. In conclusion, naturally derived antioxidant phytochemicals can be considered as future pharmaceutical drug candidates to potentially alleviate symptoms or slow the progression of PD. However, further well-designed clinical studies are required to evaluate the protective and therapeutic benefits of phytochemicals as promising drugs in the management of PD.
Collapse
Affiliation(s)
- Rengasamy Balakrishnan
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Republic of Korea
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Republic of Korea
| | - Shofiul Azam
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Republic of Korea
| | - Duk-Yeon Cho
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Republic of Korea
| | - In Su-Kim
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Republic of Korea
| | - Dong-Kug Choi
- Department of Applied Life Science, Graduate School, BK21 Program, Konkuk University, Chungju 27478, Republic of Korea
- Department of Biotechnology, College of Biomedical and Health Science, Research Institute of Inflammatory Disease (RID), Konkuk University, Chungju 27478, Republic of Korea
| |
Collapse
|
102
|
Protective Effects of p-Coumaric Acid Isolated from Vaccinium bracteatum Thunb. Leaf Extract on Corticosterone-Induced Neurotoxicity in SH-SY5Y Cells and Primary Rat Cortical Neurons. Processes (Basel) 2021. [DOI: 10.3390/pr9050869] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Corticosterone (CORT)-induced oxidative stress and neurotoxicity can cause neuronal dysfunction and mental disorders. In the present study, we investigated the effects and mechanism of the HP-20 resin fraction of the water extract of Vaccinium bracteatum leaves (NET-D1602) and its bioactive compound p-coumaric acid on neuronal cell damage in SH-SY5Y cells and primary culture of rat cortical cells. NET-D1602 and p-coumaric acid significantly improved cell viability in CORT-induced neurotoxicity in SH-SY5Y cells and primary cultures of rat cortical cells, and increased the activities of antioxidant enzymes (superoxide dismutase and catalase) against CORT-induced neurotoxicity in SH-SY5Y cells. NET-D1602 and p-coumaric acid increased the phosphorylation levels of ERK1/2 and cAMP response element-binding protein (CREB) in cortical neurons. In addition, CREB phosphorylation by NET-D1602 and p-coumaric acid was dramatically reversed by PKA, c-Raf/ERK, PI3K, and mTOR inhibitors. Lastly, we demonstrated the neuroprotective effects of NET-D1602 (3 and 10 μg/mL) and p-coumaric acid (3 and 10 μM) via increased CREB phosphorylation in CORT-induced neurotoxicity mediated via the ERK1/2, Akt, and mTOR pathways. These results suggest that p-coumaric acid is a potential neuroprotective component of NET-D1602, with the ability to protect against CORT-induced neurotoxicity by regulating ERK1/2, Akt, and mTOR-mediated CREB phosphorylation.
Collapse
|
103
|
Long HZ, Cheng Y, Zhou ZW, Luo HY, Wen DD, Gao LC. PI3K/AKT Signal Pathway: A Target of Natural Products in the Prevention and Treatment of Alzheimer's Disease and Parkinson's Disease. Front Pharmacol 2021; 12:648636. [PMID: 33935751 PMCID: PMC8082498 DOI: 10.3389/fphar.2021.648636] [Citation(s) in RCA: 251] [Impact Index Per Article: 62.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/01/2021] [Accepted: 03/08/2021] [Indexed: 12/12/2022] Open
Abstract
Alzheimer’s disease (AD) and Parkinson’s disease (PD) are two typical neurodegenerative diseases that increased with aging. With the emergence of aging population, the health problem and economic burden caused by the two diseases also increase. Phosphatidylinositol 3-kinases/protein kinase B (PI3K/AKT) signaling pathway regulates signal transduction and biological processes such as cell proliferation, apoptosis and metabolism. According to reports, it regulates neurotoxicity and mediates the survival of neurons through different substrates such as forkhead box protein Os (FoxOs), glycogen synthase kinase-3β (GSK-3β), and caspase-9. Accumulating evidences indicate that some natural products can play a neuroprotective role by activating PI3K/AKT pathway, providing an effective resource for the discovery of potential therapeutic drugs. This article reviews the relationship between AKT signaling pathway and AD and PD, and discusses the potential natural products based on the PI3K/AKT signaling pathway to treat two diseases in recent years, hoping to provide guidance and reference for this field. Further development of Chinese herbal medicine is needed to treat these two diseases.
Collapse
Affiliation(s)
- Hui-Zhi Long
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Yan Cheng
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Zi-Wei Zhou
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Hong-Yu Luo
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| | - Dan-Dan Wen
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China
| | - Li-Chen Gao
- Department of Pharmacy, Cancer Institute, Phase I Clinical Trial Centre, Changsha Central Hospital Affiliated to University of South China, School of Pharmacy, University of South China, Changsha, China.,Hunan Provincial Key Laboratory of Tumor Microenvironment Responsive Drug Research, Hengyang, China
| |
Collapse
|
104
|
Heinrich IA, Freitas AE, Wolin IAV, Nascimento APM, Walz R, Rodrigues ALS, Leal RB. Neuronal activity regulated pentraxin (narp) and GluA4 subunit of AMPA receptor may be targets for fluoxetine modulation. Metab Brain Dis 2021; 36:711-722. [PMID: 33528752 DOI: 10.1007/s11011-021-00675-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/21/2020] [Accepted: 01/22/2021] [Indexed: 12/28/2022]
Abstract
Fluoxetine is the foremost prescribed antidepressant. Drugs acting on monoaminergic system may also regulate glutamatergic system. Indeed, the investigation of proteins associated with this system, such as Narp (neuronal activity-dependent pentraxin) and GluA4 subunit of AMPA receptor may reveal poorly explored modulations triggered by conventional antidepressants. This study aimed to uncover neurochemical mechanisms underlying the chronic fluoxetine treatment, mainly by evaluating these protein targets in the prefrontal cortex and in the hippocampus. Mice received a daily administration of fluoxetine (0.1, 1 or 10 mg/kg, p.o.) or potable water (vehicle group) for 21 days. These animals were submitted to the forced swim test (FST) to verify antidepressant-like responses and the open-field test (OFT) to assess locomotor activity. Modulation of signaling proteins was analyzed by western blot. Chronic treatment with fluoxetine (1 and 10 mg/kg) was effective, since it reduced the immobility time in the FST, without altering locomotor activity. Fluoxetine 10 mg/kg increased CREB phosphorylation and BDNF expression in the prefrontal cortex and hippocampus. Noteworthy, in the hippocampus fluoxetine also promoted Akt activation and augmented Narp expression. In the prefrontal cortex, a significant decrease in the expression of the GluA4 subunit and Narp were observed following fluoxetine administration (10 mg/kg). The results provide evidence of novel molecular targets potentially involved in the antidepressant effects of fluoxetine, since in mature rodents Narp and GluA4 are mainly expressed in the GABAergic parvalbumin-positive (PV+) interneurons. This may bring new insights into the molecular elements involved in the mechanisms underlying the antidepressant effects of fluoxetine.
Collapse
Affiliation(s)
- Isabella A Heinrich
- Graduate Program in Neuroscience, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
| | - Andiara E Freitas
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
- Graduate Program in Biochemistry, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
| | - Ingrid A V Wolin
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
- Graduate Program in Biochemistry, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
| | - Ana Paula M Nascimento
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
- Graduate Program in Biochemistry, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
| | - Roger Walz
- Graduate Program in Neuroscience, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
- Department of Clinical Medicine, Center of Health Sciences, University Hospital, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
- Center of Applied Neuroscience (CeNAp), University Hospital, Federal University of Santa Catarina, Florianópolis, 88040-900, SC, Brazil
| | - Ana Lúcia S Rodrigues
- Graduate Program in Neuroscience, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
- Graduate Program in Biochemistry, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil
| | - Rodrigo B Leal
- Graduate Program in Neuroscience, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil.
- Department of Biochemistry, Center of Biological Sciences, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil.
- Graduate Program in Biochemistry, Federal University of Santa Catarina, Campus Universitário, Trindade, Florianópolis, 88040-900, SC, Brazil.
| |
Collapse
|
105
|
Moore NS, Mans RA, McCauley MK, Allgood CS, Barksdale KA. Critical Effects on Akt Signaling in Adult Zebrafish Brain Following Alterations in Light Exposure. Cells 2021; 10:cells10030637. [PMID: 33809219 PMCID: PMC8000057 DOI: 10.3390/cells10030637] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/31/2021] [Revised: 03/08/2021] [Accepted: 03/09/2021] [Indexed: 11/16/2022] Open
Abstract
Evidence from human and animal studies indicate that disrupted light cycles leads to alterations of the sleep state, poor cognition, and the risk of developing neuroinflammatory and generalized health disorders. Zebrafish exhibit a diurnal circadian rhythm and are an increasingly popular model in studies of neurophysiology and neuropathophysiology. Here, we investigate the effect of alterations in light cycle on the adult zebrafish brain: we measured the effect of altered, unpredictable light exposure in adult zebrafish telencephalon, homologous to mammalian hippocampus, and the optic tectum, a significant visual processing center with extensive telencephalon connections. The expression of heat shock protein-70 (HSP70), an important cell stress mediator, was significantly decreased in optic tectum of adult zebrafish brain following four days of altered light exposure. Further, pSer473-Akt (protein kinase B) was significantly reduced in telencephalon following light cycle alteration, and pSer9-GSK3β (glycogen synthase kinase-3β) was significantly reduced in both the telencephalon and optic tectum of light-altered fish. Animals exposed to five minutes of environmental enrichment showed significant increase in pSer473Akt, which was significantly attenuated by four days of altered light exposure. These data show for the first time that unpredictable light exposure alters HSP70 expression and dysregulates Akt-GSK3β signaling in the adult zebrafish brain.
Collapse
|
106
|
Zeng C, Yang P, Cao T, Gu Y, Li N, Zhang B, Xu P, Liu Y, Luo Z, Cai H. Gut microbiota: An intermediary between metabolic syndrome and cognitive deficits in schizophrenia. Prog Neuropsychopharmacol Biol Psychiatry 2021; 106:110097. [PMID: 32916223 DOI: 10.1016/j.pnpbp.2020.110097] [Citation(s) in RCA: 20] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 08/29/2020] [Accepted: 09/01/2020] [Indexed: 12/12/2022]
Abstract
Gut microbiome interacts with the central nervous system tract through the gut-brain axis. Such communication involves neuronal, endocrine, and immunological mechanisms, which allows for the microbiota to affect and respond to various behaviors and psychiatric conditions. In addition, the use of atypical antipsychotic drugs (AAPDs) may interact with and even change the abundance of microbiome to potentially cause adverse effects or aggravate the disorders inherent in the disease. The regulate effects of gut microbiome has been described in several psychiatric disorders including anxiety and depression, but only a few reports have discussed the role of microbiota in AAPDs-induced Metabolic syndrome (MetS) and cognitive disorders. The following review systematically summarizes current knowledge about the gut microbiota in behavior and psychiatric illness, with the emphasis of an important role of the microbiome in the metabolism of schizophrenia and the potential for AAPDs to change the gut microbiota to promote adverse events. Prebiotics and probiotics are microbiota-management tools with documented efficacy for metabolic disturbances and cognitive deficits. Novel therapies for targeting microbiota for alleviating AAPDs-induced adverse effects are also under fast development.
Collapse
Affiliation(s)
- CuiRong Zeng
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, China; The Institute of Clinical Pharmacy, Central South University, Changsha 410011, Hunan Province, China
| | - Ping Yang
- Department of Psychiatry, The Second People's Hospital of Hunan Province, Changsha 410007, Hunan Province, China
| | - Ting Cao
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, China; The Institute of Clinical Pharmacy, Central South University, Changsha 410011, Hunan Province, China
| | - YuXiu Gu
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, China; The Institute of Clinical Pharmacy, Central South University, Changsha 410011, Hunan Province, China
| | - NaNa Li
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, China; The Institute of Clinical Pharmacy, Central South University, Changsha 410011, Hunan Province, China
| | - BiKui Zhang
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, China; The Institute of Clinical Pharmacy, Central South University, Changsha 410011, Hunan Province, China
| | - Ping Xu
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, China; The Institute of Clinical Pharmacy, Central South University, Changsha 410011, Hunan Province, China
| | - YiPing Liu
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, China; The Institute of Clinical Pharmacy, Central South University, Changsha 410011, Hunan Province, China
| | - ZhiYing Luo
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, China; The Institute of Clinical Pharmacy, Central South University, Changsha 410011, Hunan Province, China
| | - HuaLin Cai
- Department of Pharmacy, The Second Xiangya Hospital of Central South University, Changsha 410011, Hunan Province, China; The Institute of Clinical Pharmacy, Central South University, Changsha 410011, Hunan Province, China.
| |
Collapse
|
107
|
Zhong W, Darmani NA. Role of PI3K/Akt/GSK-3 Pathway in Emesis and Potential New Antiemetics. JOURNAL OF CELLULAR SIGNALING 2020; 1:155-159. [PMID: 33426544 PMCID: PMC7793561 DOI: 10.33696/signaling.1.024] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Key Words] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- W Zhong
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 East Second Street, Pomona, CA 91766, USA
| | - N A Darmani
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 East Second Street, Pomona, CA 91766, USA
| |
Collapse
|
108
|
Laviolette SR. Molecular and neuronal mechanisms underlying the effects of adolescent nicotine exposure on anxiety and mood disorders. Neuropharmacology 2020; 184:108411. [PMID: 33245960 DOI: 10.1016/j.neuropharm.2020.108411] [Citation(s) in RCA: 27] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2020] [Revised: 11/16/2020] [Accepted: 11/21/2020] [Indexed: 12/28/2022]
Abstract
Tobacco addiction is highly co-morbid with a variety of mental health conditions, including schizophrenia, mood and anxiety disorders. Nicotine, the primary psychoactive compound in tobacco-related products is known to functionally modulate brain circuits that are disturbed in these disorders. Nicotine can potently regulate the transmission of various neurochemicals, including dopamine (DA), γ-amino-butyric acid (GABA) and glutamate, within various mesocorticolimbic structures, such as the ventral tegmental area (VTA), nucleus accumbens (NAc) and prefrontal cortex (PFC), all of which show pathologies in these disorders. Many neuropsychiatric diseases have etiological origins during neurodevelopment, typically occurring during vulnerable periods of adolescent or pre-natal brain development. During these neurodevelopmental periods, exposure to extrinsic drug insults can induce enduring and long-term pathophysiological sequelae that ultimately increase the risk of developing chronic mental health disorders in later life. These vulnerability factors are of growing concern given rising rates of adolescent nicotine exposure via traditional tobacco use and the increasing use of alternative nicotine delivery formats such as vaping and e-cigarettes. A large body of clinical and pre-clinical evidence points to an important role for adolescent exposure to nicotine and increased vulnerability to developing mood and anxiety disorders in later life. This review will examine current clinical and pre-clinical evidence that pinpoints specific mechanisms within the mesocorticolimbic circuitry and molecular biomarkers linked to the association between adolescent nicotine exposure and increased risk of developing mood and anxiety-related disorders. This article is part of the special issue on 'Vulnerabilities to Substance Abuse'.
Collapse
Affiliation(s)
- Steven R Laviolette
- Addiction Research Group, Dept. of Anatomy & Cell Biology, Dept. of Psychiatry, Schulich School of Medicine and Dentistry, University of Western Ontario, London, N6A 3K7, ON, Canada.
| |
Collapse
|
109
|
Sargazi S, Heidari Nia M, Sheervalilou R, Mirinejad S, Harati-Sadegh M, Moudi M, Saravani R, Shakiba M. Relationship between Single Nucleotide Polymorphisms of GRHL3 and Schizophrenia Susceptibility: A Preliminary Case-Control Study and Bioinformatics Analysis. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2020; 9:154-164. [PMID: 32934953 PMCID: PMC7489109 DOI: 10.22088/ijmcm.bums.9.2.154] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 07/28/2020] [Indexed: 12/28/2022]
Abstract
Grainyhead-like (GRHL) transcription factors were recently linked to the etiology of neural tube defects (NTDs). Overlapping patterns in the variation of schizophrenia (SCZ) incidence with that of NTDs suggests the presence of common etiological risk factors. This preliminary study was designed to examine the relationship between two missense variants of GRHL3 gene (rs2486668C/G and rs545809A/T) and SCZ susceptibility among Iranians. Three hundred ninety subjects (192 patients confirmed with SCZ, and 198 healthy controls) were enrolled and genotyped. Statistical and bioinformatics analyzes were performed to determine the effects of the variants. In silico analyzes were performed to determine the effects of the variants on the secondary structure of GRHL3 protein and prediction of silencer motifs for each variation. Statistically significant differences were observed between the studied groups under codominant AA, dominant AT+AA, and recessive AA genetic contrast models for rs545809A/T. The presence of the A allele of rs545809A/T enhanced SCZ risk by 2.33 fold. In contrast, rs2486668C/G was not linked to SCZ susceptibility (P > 0.05). Bioinformatics analysis revealed that both missense SNPs caused substantial changes in the secondary structure of GRHL3-mRNA. Screening of the flanking sequences of rs545809A/T predicted silencer motifs for this SNP. Our results demonstrated that the rs545809A/T of GRHL3 gene could affect the risk of SCZ in Iranian populations. Replication studies are warranted to confirm these results.
Collapse
Affiliation(s)
- Saman Sargazi
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Milad Heidari Nia
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Roghayeh Sheervalilou
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Shekoufeh Mirinejad
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mahdiyeh Harati-Sadegh
- Genetics of Non-Communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mahdiyeh Moudi
- Genetics of Non-Communicable Disease Research Center, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Ramin Saravani
- Cellular and Molecular Research Center, Resistant Tuberculosis Institute, Zahedan University of Medical Sciences, Zahedan, Iran.,Department of Clinical Biochemistry, School of Medicine, Zahedan University of Medical Sciences, Zahedan, Iran
| | - Mansoor Shakiba
- Department of Psychiatry, Zahedan University of Medical Sciences, Zahedan, Iran.,Psychosomatic Research Center, Imam Hospital, Tehran University of Medical Sciences, Tehran, Iran
| |
Collapse
|
110
|
Zhao F, Tao W, Shang Z, Zhang W, Ruan J, Zhang C, Zhou L, Aiello H, Lai H, Qu R. Facilitating Granule Cell Survival and Maturation in Dentate Gyrus With Baicalin for Antidepressant Therapeutics. Front Pharmacol 2020; 11:556845. [PMID: 32982755 PMCID: PMC7493074 DOI: 10.3389/fphar.2020.556845] [Citation(s) in RCA: 23] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2020] [Accepted: 08/18/2020] [Indexed: 01/02/2023] Open
Abstract
Baicalin isolated from Scutellaria baicalensis possesses antidepressant abilities through its relation to hippocampal neurogenesis. Current research has found that baicalin can promote the proliferation of hippocampal granule cells, however, the detailed mechanism of baicalin on the survival and maturation of hippocampal granule cells has yet to be sufficiently explored. The purpose of this study was to evaluate whether baicalin could facilitate the survival and maturation of hippocampal granule cells, and to explore its potential mechanism. The chronic corticosterone (CORT)-induced mouse model of depression was used to assess antidepressant-like effects of baicalin and to illuminate possible molecular mechanisms by which baicalin affects hippocampal neurogenesis. The survival and maturation of granule cells were measured by immunohistochemistry, immunofluorescence and Golgi staining. The expression of Phosphatidylinositol 3-kinase (PI3K)/Protein kinase B (AKT)/glycogen synthase kinase-3β (GSK3β)/β-catenin pathway related proteins were measured by western blot analysis. PI3K inhibitor LY292002 and AKT inhibitor Perifosine were administered to HT-22 cells to explore the relationship between the PI3K/AKT/GSK3β/β-catenin pathway and baicalin. The results of the study illustrated that baicalin significantly decreased chronic CORT-induced depressive-like behaviors and reduced serum corticosterone levels. In addition, baicalin (administered at 60 mg/kg) reversed chronic CORT-induced lesions on hippocampal granule cells. Moreover, baicalin significantly increased the phosphorylation rate of PI3K, AKT, GSK3β, and total β-catenin. The study found that administration of LY292002/Perifosine counteracted the effects of baicalin in HT-22 cells. These results demonstrate that baicalin can alleviate chronic CORT-induced depressive-like behaviors through promoting survival and maturation of adult-born hippocampal granule cells and exhibiting protective effect on hippocampal neuron morphology. We propose the underlying mechanisms involve the activation of the PI3K/AKT/GSK3β/β-catenin pathway.
Collapse
Affiliation(s)
- Fan Zhao
- College of Chinese Medicine, College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weiwei Tao
- College of Chinese Medicine, College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Zhiyuan Shang
- College of Chinese Medicine, College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Weihua Zhang
- College of Chinese Medicine, College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Jie Ruan
- College of Chinese Medicine, College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Chenyiyu Zhang
- College of Chinese Medicine, College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | - Liping Zhou
- College of Chinese Medicine, College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| | | | - Hezheng Lai
- Chinese Medicine Centre, Western Sydney University, Campbelltown, NSW, Australia.,NICM Health Research Institute, Western Sydney University, Westmead, NSW, Australia
| | - Rong Qu
- College of Chinese Medicine, College of Integrated Chinese and Western Medicine, Nanjing University of Chinese Medicine, Nanjing, China
| |
Collapse
|
111
|
Watkins LR, Orlandi C. Orphan G Protein Coupled Receptors in Affective Disorders. Genes (Basel) 2020; 11:E694. [PMID: 32599826 PMCID: PMC7349732 DOI: 10.3390/genes11060694] [Citation(s) in RCA: 37] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2020] [Revised: 06/20/2020] [Accepted: 06/21/2020] [Indexed: 12/12/2022] Open
Abstract
G protein coupled receptors (GPCRs) are the main mediators of signal transduction in the central nervous system. Therefore, it is not surprising that many GPCRs have long been investigated for their role in the development of anxiety and mood disorders, as well as in the mechanism of action of antidepressant therapies. Importantly, the endogenous ligands for a large group of GPCRs have not yet been identified and are therefore known as orphan GPCRs (oGPCRs). Nonetheless, growing evidence from animal studies, together with genome wide association studies (GWAS) and post-mortem transcriptomic analysis in patients, pointed at many oGPCRs as potential pharmacological targets. Among these discoveries, we summarize in this review how emotional behaviors are modulated by the following oGPCRs: ADGRB2 (BAI2), ADGRG1 (GPR56), GPR3, GPR26, GPR37, GPR50, GPR52, GPR61, GPR62, GPR88, GPR135, GPR158, and GPRC5B.
Collapse
Affiliation(s)
| | - Cesare Orlandi
- Department of Pharmacology and Physiology, University of Rochester Medical Center, Rochester, NY 14642, USA;
| |
Collapse
|
112
|
Vinay P, Karen C, Balamurugan K, Rajan KE. Cronobacter sakazakii Infection in Early Postnatal Rats Impaired Contextual-Associated Learning: a Putative Role of C5a-Mediated NF-κβ and ASK1 Pathways. J Mol Neurosci 2020; 71:28-41. [PMID: 32567007 DOI: 10.1007/s12031-020-01622-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2019] [Accepted: 06/08/2020] [Indexed: 12/28/2022]
Abstract
This study was designed to test whether the Cronobacter sakazakii infection-impaired contextual learning and memory are mediated by the activation of the complement system; subsequent activation of inflammatory signals leads to alternations in serotonin transporter (SERT). To test this, rat pups (postnatal day, PND 15) were treated with either C. sakazakii (107 CFU) or Escherichia coli OP50 (107 CFU) or Luria bertani broth (100 μL) through oral gavage and allowed to stay with their mothers until PND 24. Experimental groups' rats were allowed to explore (PNDs 31-35) and then trained in contextual learning task (PNDs 36-43). Five days after training, individuals were tested for memory retention (PNDs 49-56). Observed behavioural data showed that C. sakazakii infection impaired contextual-associative learning and memory. Furthermore, our analysis showed that C. sakazakii infection activates complement system complement anaphylatoxin (C5a) (a disintegrin and metalloproteinase with thrombospondin motifs (ADAMTS1)) and mitogen-activated protein kinase kinase1 (MEKK1). Subsequently, MEKK1 induces pro-inflammatory signals possibly through apoptosis signal-regulating kinase-1 (ASK-1), c-Jun N-terminal kinase (JNK1/3) and protein kinase B gamma (AKT-3). In parallel, activated nuclear factor kappa-light-chain-enhancer B cells (NF-κB) induces interleukin-6 (IL-6) and IFNα-1, which may alter the level of serotonin transporter (SERT). Observed results suggest that impaired contextual learning and memory could be correlated with C5a-mediated NF-κβ and ASK1 pathways.
Collapse
Affiliation(s)
- Ponnusamy Vinay
- Behavioural Neuroscience Laboratory, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, India
| | - Christopher Karen
- Behavioural Neuroscience Laboratory, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, India
| | | | - Koilmani Emmanuvel Rajan
- Behavioural Neuroscience Laboratory, Department of Animal Science, School of Life Sciences, Bharathidasan University, Tiruchirappalli, 620024, India.
| |
Collapse
|
113
|
Murakami M, Ikeda Y, Nakagawa Y, Tsuji A, Kitagishi Y, Matsuda S. Special bioactive compounds and functional foods may exhibit neuroprotective effects in patients with dementia (Review). Biomed Rep 2020; 13:1. [PMID: 32509304 PMCID: PMC7271706 DOI: 10.3892/br.2020.1310] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2019] [Accepted: 05/07/2020] [Indexed: 12/13/2022] Open
Abstract
Dementia is a failure of cognitive ability characterized by severe neurodegeneration in select neural systems, and Alzheimer's disease (AD) is the most common type of neurodegenerative disease. Although numerous studies have provided insights into the pathogenesis of AD, the underlying signaling and molecular pathways mediating the progressive decline of cognitive function remain poorly understood. Recent progress in molecular biology has provided an improved understanding of the importance of molecular pathogenesis of AD, and has proposed an association between DNA repair mechanisms and AD. In particular, the fundamental roles of phosphatase and tensin homologue deleted on chromosome 10 (PTEN) and breast cancer gene 1 (BRCA1) tumor suppressors have been shown to regulate the pathogenesis of neurodegeneration. Consequently, onset of neurodegenerative diseases may be deferred with the use of dietary neuroprotective agents which alter the signaling mediated by the aforementioned tumor suppressors. In a healthy neuron, homeostasis of key intracellular molecules is of great importance, and preventing neuronal apoptosis is one of the primary goals of treatments designed for dementia-associated diseases. In the present review, progress into the understanding of dietary regulation for preventing or limiting development of dementia is discussed with a focus on the modulatory roles of PTEN and BRCA1 signaling.
Collapse
Affiliation(s)
- Mutsumi Murakami
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Yuka Ikeda
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Yukie Nakagawa
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Ai Tsuji
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Yasuko Kitagishi
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| | - Satoru Matsuda
- Department of Food Science and Nutrition, Nara Women's University, Kita-Uoya Nishimachi, Nara 630-8506, Japan
| |
Collapse
|
114
|
Kobayashi Y, Takeda T, Kunitomi H, Ueki A, Misu K, Kowashi A, Takahashi T, Anko M, Watanabe K, Masuda K, Uchida T, Tominaga E, Banno K, Kosaki K, Aoki D. Cowden syndrome complicated by schizophrenia: A first clinical report. Eur J Med Genet 2020; 63:103959. [PMID: 32461083 DOI: 10.1016/j.ejmg.2020.103959] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Revised: 04/28/2020] [Accepted: 05/17/2020] [Indexed: 11/27/2022]
Abstract
Patients with Cowden syndrome exhibit mucocutaneous lesions, hamartomatous polyposis of the gastrointestinal tract, and macrocephaly, often complicated by malignant tumors, such as breast, thyroid, and uterine cancers. Autism spectrum and epilepsy have been known as neuropsychiatric symptoms associated with Cowden syndrome; however, to the best of our knowledge, there is no report on cases complicated by schizophrenia. Here, we report a first case of Cowden syndrome complicated by schizophrenia. A 49-year-old Japanese woman started experiencing auditory hallucinations in her teens. She had left breast cancer at the age of 34 years, and right breast cancer at the age of 37 years, all of which were surgically treated. She was also being treated by oral medications for Hashimoto's disease. On consulting her previous doctor for abnormal uterine bleeding that lasted for a year, she was diagnosed with endometrial cancer. However, immediately before surgery, her auditory hallucinations and paranoid delusions became severe, and she was referred to our hospital for detailed examination and treatment. No abnormalities were found on head MRI, and she was diagnosed with schizophrenia on the basis of neuropsychiatric examination findings. After her psychiatric symptoms were controlled by 2 mg of risperidone, she underwent surgery for endometrial cancer. Although there was no apparent family history, physical findings including macrocephaly and papillomatous skin lesions together with her past medical history of multiple malignant tumors suggested Cowden syndrome. Postoperatively, genetic testing revealed a pathogenic variant c.655C > T; p. Gln219* (NM_000314.4) in PTEN, leading to the confirmation of the diagnosis of Cowden syndrome.
Collapse
Affiliation(s)
- Yusuke Kobayashi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan; Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan.
| | - Takashi Takeda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Haruko Kunitomi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Arisa Ueki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Kumiko Misu
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Ayari Kowashi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Takayuki Takahashi
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Mayuka Anko
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Keiko Watanabe
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kenta Masuda
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Takahito Uchida
- Department of Neuropsychiatry, Keio University School of Medicine, Tokyo, Japan
| | - Eiichiro Tominaga
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kouji Banno
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| | - Kenjiro Kosaki
- Center for Medical Genetics, Keio University School of Medicine, Tokyo, Japan
| | - Daisuke Aoki
- Department of Obstetrics and Gynecology, Keio University School of Medicine, Tokyo, Japan
| |
Collapse
|
115
|
Dela Justina V, Priviero F, Dos Passos RR, Webb RC, Lima VV, Giachini FR. O-GlcNAc impairs endothelial function in uterine arteries from virgin but not pregnant rats: The role of GSK3β. Eur J Pharmacol 2020; 880:173133. [PMID: 32343970 DOI: 10.1016/j.ejphar.2020.173133] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2020] [Revised: 04/18/2020] [Accepted: 04/20/2020] [Indexed: 01/10/2023]
Abstract
Increased O-Linked β-N-acetylglucosamine (O-GlcNAc) is observed in several pathologies, and unbalanced O-GlcNAcylation levels favor endothelial dysfunction. Whether augmented O-GlcNAc impacts the uterine artery (UA) function and how it affects the UA during pregnancy remains to be elucidated. We hypothesized that glucosamine treatment increases O-GlcNAc, leading to uterine artery dysfunction and this effect is prevented by pregnancy. Pregnant (P) and non-pregnant (NP) Wistar rats were treated with glucosamine (300 mg/kg; i.p.) for 21 days. Concentration response-curves (CRC) to acetylcholine (in the presence or absence of L-NAME) and sodium nitroprusside were performed in UAs. In NP rats, glucosamine treatment increased O-GlcNAc expression in UAs accompanied by decreased endothelium-dependent relaxation, which was abolished by L-NAME. Endothelial nitric oxide synthase (eNOS) activity and total Akt expression were decreased by glucosamine-treatment in NP rats. Further, NP rats treated with glucosamine displayed increased glycogen synthase kinase 3 beta (GSK3β) activation and O-GlcNAc-transferase (OGT) expression in the UA. P rats treated with glucosamine displayed decreased O-GlcNAc in UAs and it was accompanied by improved relaxation to acetylcholine, whereas eNOS and GSK3β activity and total Akt and OGT expression were unchanged. Sodium nitroprusside-induced relaxation was not changed in all groups, indicating that glucosamine treatment led to endothelial dysfunction in NP rats. The underlying mechanism is, at least in part, dependent on Akt/GSK3β/OGT modulation. We speculate that during pregnancy, hormonal alterations play a protective role in preventing O-GlcNAcylation-induced endothelial dysfunction in the UAs.
Collapse
Affiliation(s)
- Vanessa Dela Justina
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil
| | | | - Rinaldo Rodrigues Dos Passos
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil; Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, MT, Brazil
| | - R Clinton Webb
- Department of Physiology, Augusta University, Augusta, GA, USA
| | - Victor Vitorino Lima
- Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, MT, Brazil
| | - Fernanda R Giachini
- Graduate Program in Biological Sciences, Federal University of Goiás, Goiânia, GO, Brazil; Institute of Biological Sciences and Health, Federal University of Mato Grosso, Barra do Garças, MT, Brazil.
| |
Collapse
|
116
|
Xu F, Na L, Li Y, Chen L. Roles of the PI3K/AKT/mTOR signalling pathways in neurodegenerative diseases and tumours. Cell Biosci 2020; 10:54. [PMID: 32266056 PMCID: PMC7110906 DOI: 10.1186/s13578-020-00416-0] [Citation(s) in RCA: 389] [Impact Index Per Article: 77.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2019] [Accepted: 03/26/2020] [Indexed: 02/06/2023] Open
Abstract
The PI3 K/AKT/mTOR signalling pathway plays an important role in the regulation of signal transduction and biological processes such as cell proliferation, apoptosis, metabolism and angiogenesis. Compared with those of other signalling pathways, the components of the PI3K/AKT/mTOR signalling pathway are complicated. The regulatory mechanisms and biological functions of the PI3K/AKT/mTOR signalling pathway are important in many human diseases, including ischaemic brain injury, neurodegenerative diseases, and tumours. PI3K/AKT/mTOR signalling pathway inhibitors include single-component and dual inhibitors. Numerous PI3K inhibitors have exhibited good results in preclinical studies, and some have been clinically tested in haematologic malignancies and solid tumours. In this review, we briefly summarize the results of research on the PI3K/AKT/mTOR pathway and discuss the structural composition, activation, communication processes, regulatory mechanisms and biological functions of the PI3K/AKT/mTOR signalling pathway in the pathogenesis of neurodegenerative diseases and tumours.
Collapse
Affiliation(s)
- Fei Xu
- Department of Microbiology and Immunology, Shanghai University of Medicine & Health Sciences, 279 Zhouzhu Rd, Shanghai, 201318 China
- Collaborative Innovation Center of Shanghai University of Medicine & Health Sciences, Shanghai, 201318 China
| | - Lixin Na
- Collaborative Innovation Center of Shanghai University of Medicine & Health Sciences, Shanghai, 201318 China
- Department of Inspection and Quarantine, Shanghai University of Medicine & Health Sciences, Shanghai, 201318 China
| | - Yanfei Li
- Department of Inspection and Quarantine, Shanghai University of Medicine & Health Sciences, Shanghai, 201318 China
| | - Linjun Chen
- Department of Inspection and Quarantine, Shanghai University of Medicine & Health Sciences, Shanghai, 201318 China
| |
Collapse
|
117
|
Chuang HW, Wei IH, Lin FY, Li CT, Chen KT, Tsai MH, Huang CC. Roles of Akt and ERK in mTOR-Dependent Antidepressant Effects of Vanillic Acid. ACS OMEGA 2020; 5:3709-3716. [PMID: 32118186 PMCID: PMC7045503 DOI: 10.1021/acsomega.9b04271] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2019] [Accepted: 01/31/2020] [Indexed: 05/05/2023]
Abstract
Vanillic acid, an oxidized form of vanilla, is a flavoring agent with a creamy odor. Several studies have reported the neuroprotective effects of vanillic acid, which are predominantly associated with anti-inflammatory and antioxidative properties. The anti-inflammatory and antioxidative properties may result from Akt or ERK signaling activation. The activation of the mammalian target of rapamycin (mTOR), a key downstream target of Akt and ERK signaling, is a crucial therapeutic target for treating depression. However, the antidepressant effects of vanillic acid remain unknown. The present study applied the forced swim test (FST) to investigate the antidepressant effects of vanillic acid and its association with Akt, ERK, and mTOR signaling and upstream α-amino-3-hydroxy-5-methyl-4-isoxazolepropionaic acid receptor (AMPAR) in the prefrontal cortex (PFC) of mice. Vanillic acid demonstrated antidepressant effects by significantly reducing behavioral despair in the FST. None of the treatments changed locomotor activity. Additionally, vanillic acid increased AMPAR throughput, Akt, and mTOR signaling but not ERK signaling in the PFC. NBQX (an AMPAR blocker), MK 2206 (an Akt blocker), and rapamycin (an mTOR blocker) used in pretreatment attenuated the antidepressant effects of vanillic acid, but SL327 (an ERK inhibitor) did not. The immunochemical results indicated that the antidepressant effects of vanillic acid depend on the AMPAR-Akt-mTOR signaling transduction pathway. Our findings reveal an Akt-dependent, but ERK-independent, the mechanism underlying the antidepressant effects of vanillic acid, which may be beneficial for some patients with depression.
Collapse
Affiliation(s)
- Han-Wen Chuang
- Graduate Institute
of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
| | - I-Hua Wei
- Department of Anatomy, China Medical University, Taichung 40402, Taiwan
| | - Fang-Yi Lin
- Department of Medical Laboratory Science and Biotechnology, China Medical University, Taichung 40402, Taiwan
| | - Chun-Te Li
- Department of Medicine, China
Medical University, Taichung 40402, Taiwan
| | - Kuang-Ti Chen
- Department
of Veterinary Medicine, National Chung Hsing
University, Taichung 40227, Taiwan
| | - Mang-Hung Tsai
- Department of Anatomy, China Medical University, Taichung 40402, Taiwan
| | - Chih-Chia Huang
- Graduate Institute
of Biomedical Sciences, China Medical University, Taichung 40402, Taiwan
- Department of
Psychiatry, China Medical University, Taichung 40402, Taiwan
- Department of Psychiatry, China Medical University Hospital, Taichung 40447, Taiwan
- E-mail: . Tel: 886-4-22052121 ext 1015. Fax: 886-4-22361230
| |
Collapse
|
118
|
Ceylan D, Tufekci KU, Keskinoglu P, Genc S, Özerdem A. Circulating exosomal microRNAs in bipolar disorder. J Affect Disord 2020; 262:99-107. [PMID: 31726266 DOI: 10.1016/j.jad.2019.10.038] [Citation(s) in RCA: 36] [Impact Index Per Article: 7.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/08/2019] [Revised: 09/23/2019] [Accepted: 10/27/2019] [Indexed: 12/20/2022]
Abstract
INTRODUCTION Emerging evidence suggests central roles of miRNAs in the pathogenesis of bipolar disorder (BD). Exosomes are membrane-bound vesicles acting as "biological cargo carriers" of various types of molecules including microRNAs. In this study, we aimed to investigate circulating exosomal microRNAs as potential diagnostic biomarkers for BD. METHODS The exosomes were precipitated from plasma samples of patients with BD (n = 69; 15 depressed, 27 manic, 27 euthymic) and healthy controls (n = 41). Total RNA was extracted from the exosomes and the levels of miRNAs were assayed by qPCR. Dysregulated miRNAs were subjected to Kyoto Encyclopedia of Genes and Genomes" (KEGG) pathway analysis by DIANA-miRPath v3.0 to identify the predicted targets and the related pathways. RESULTS Thirteen miRNAs showed significant differences between patients with BD and healthy individuals; among these, MiR-484, -652-3p, -142-3p remained significantly downregulated and miR-185-5p remained significantly upregulated after accounting for multiple comparisons and adjustments for potential confounders. There were no significant alterations among different states of BD. The KEEG analysis of four dysregulated miRNAs highlighted several target pathways including PI3K/Akt signaling, fatty acid biosynthesis/metabolism, extracellular matrix and adhesion pathways. CONCLUSION Our findings suggest that dysregulation of miRNAs might be involved in the underlying pathophysiology of BD through several biological pathways; and highlight the importance of the exosomal miRNAs for biomarker research in BD. Further longitudinal studies may clarify the roles of exosomal miRNAs and their targets in the neurobiology of BD.
Collapse
Affiliation(s)
- Deniz Ceylan
- Izmir University of Economics, Faculty of Medicine, Department of Psychiatry, Izmir, Turkey
| | - Kemal Ugur Tufekci
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Pembe Keskinoglu
- Department of Biostatistics and Medical Informatics, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey
| | - Sermin Genc
- Izmir Biomedicine and Genome Center, Dokuz Eylul University Health Campus, Izmir, Turkey; Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University Health Campus, Izmir, Turkey
| | - Ayşegül Özerdem
- Department of Neuroscience, Institute of Health Sciences, Dokuz Eylul University Health Campus, Izmir, Turkey; Department of Psychiatry and Psychology, Mayo Clinic, Rochester, MN, USA; Department of Psychiatry, Dokuz Eylul University Faculty of Medicine, Izmir, Turkey.
| |
Collapse
|
119
|
NOX2-Dependent Reactive Oxygen Species Regulate Formyl-Peptide Receptor 1-Mediated TrkA Transactivation in SH-SY5Y Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2019; 2019:2051235. [PMID: 31871542 PMCID: PMC6913242 DOI: 10.1155/2019/2051235] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 07/11/2019] [Revised: 10/08/2019] [Accepted: 10/18/2019] [Indexed: 12/16/2022]
Abstract
Several enzymes are capable of producing reactive oxygen species (ROS), but only NADPH oxidases (NOX) generate ROS as their primary and sole function. In the central nervous system, NOX2 is the major source of ROS, which play important roles in signalling and functions. NOX2 activation requires p47phox phosphorylation and membrane translocation of cytosolic subunits. We demonstrate that SH-SY5Y cells express p47phox and that the stimulation of Formyl-Peptide Receptor 1 (FPR1) by N-fMLP induces p47phox phosphorylation and NOX-dependent superoxide generation. FPR1 is a member of the G protein-coupled receptor (GPCR) family and is able to transphosphorylate several tyrosine kinase receptors (RTKs). This mechanism requires ROS as signalling intermediates and is necessary to share information within the cell. We show that N-fMLP stimulation induces the phosphorylation of cytosolic Y490, Y751, and Y785 residues of the neurotrophin receptor TrkA. These phosphotyrosines provide docking sites for signalling molecules which, in turn, activate Ras/MAPK, PI3K/Akt, and PLC-γ1/PKC intracellular cascades. N-fMLP-induced ROS generation plays a critical role in FPR1-mediated TrkA transactivation. In fact, the blockade of NOX2 functions prevents Y490, Y751, and Y785 phosphorylation, as well as the triggering of downstream signalling cascades. Moreover, we observed that FPR1 stimulation by N-fMLP also improves proliferation, cellular migration, and neurite outgrowth of SH-SY5Y cells.
Collapse
|
120
|
Wang H, Xiao L, Wang H, Wang G. Involvement of chronic unpredictable mild stress-induced hippocampal LRP1 up-regulation in microtubule instability and depressive-like behavior in a depressive-like adult male rat model. Physiol Behav 2019; 215:112749. [PMID: 31770536 DOI: 10.1016/j.physbeh.2019.112749] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2019] [Revised: 11/07/2019] [Accepted: 11/22/2019] [Indexed: 12/18/2022]
Abstract
Low-density lipoprotein receptor-related protein 1 (LRP1) and tau play an important role in developing Alzheimer's disease. This study aimed to explore the involvement of LRP1 in microtubule dynamic and depressive-like behavior in a depressive-like rat model. It also investigated whether fluoxetine blocked the change induced by chronic unpredictable mild stress (CUMS). Sprague-Dawley rats (200-250 g) were exposed to CUMS and fluoxetine for 4 weeks respectively. The body weight was determined, and behavior tests, including sucrose preference test, forced swimming test and open field test were performed. Western blot analysis was conducted to determine the protein levels of LRP1, tubulin, Acet-tub, Tyr-tub and PI3K/Akt/GSK-3β. Real-time quantitative polymerase chain reaction was used for mRNA expression levels of LRP1. Immunohistochemical staining was applied for LRP1 and immunofluorescence staining for the co-location of p-tau (404,262) and Acet-tub. The CUMS group presented a decreased body weight and depressive-like behavior, which was improved by fluoxetine. The protein and mRNA expression levels of LRP1 were elevated in the CUMS group. The levels of Acet-tub increased following CUMS, accompanied by elevated levels of p-tau (404,262). The binding of p-tau and Acet-tub significantly decreased in depressive-like rats, and fluoxetine attenuated microtubule instability. Finally, the inhibition of CUMS-induced PI3K/Akt activated GSK-3β, and fluoxetine reversed the change in the signaling pathway. Hence, LRP1 might impair the microtubule dynamics accompanied by depressive-like behavior via the PI3K/ Akt /GSK3β pathway in adult depressive-like rats, and hippocampal LRP1 might be involved in the development of depression.
Collapse
Affiliation(s)
- Hui Wang
- Department of Psychiatry, Renmin hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China
| | - Ling Xiao
- Department of Psychiatry, Renmin hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China
| | - Huiling Wang
- Department of Psychiatry, Renmin hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China
| | - Gaohua Wang
- Department of Psychiatry, Renmin hospital of Wuhan University, Jiefang Road 238#, Wuhan 430060, Hubei, PR China.
| |
Collapse
|
121
|
Zhong W, Darmani NA. The pivotal role of glycogen synthase kinase 3 (GSK-3) in vomiting evoked by specific emetogens in the least shrew (Cryptotis parva). Neurochem Int 2019; 132:104603. [PMID: 31738972 DOI: 10.1016/j.neuint.2019.104603] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/26/2019] [Revised: 11/08/2019] [Accepted: 11/12/2019] [Indexed: 12/27/2022]
Abstract
Glycogen synthase kinase 3 (GSK-3) is a constitutively active multifunctional serine-threonine kinase which is involved in diverse physiological processes. GSK-3 has been implicated in a wide range of diseases including neurodegeneration, inflammation, diabetes and cancer. GSK-3 is a downstream target for protein kinase B (Akt) which phosphorylates GSK-3 and suppresses its activity. Based upon our preliminary findings, we postulated Akt's involvement in emesis. The aim of this study was to investigate the participation of GSK-3 and the antiemetic potential of two GSK-3 inhibitors (AR-A014418 and SB216763) in the least shrew model of vomiting against fully-effective emetic doses of diverse emetogens, including the nonselective and/or selective agonists of serotonin type 3 (e.g. 5-HT or 2-Methyl-5-HT)-, neurokinin type 1 receptor (e.g. GR73632), dopamine D2 (e.g. apomorphine or quinpirole)-, and muscarinic 1 (e.g. pilocarpine or McN-A-343) receptors, as well as the L-type Ca2+ channel agonist (FPL64176), the sarco/endoplasmic reticulum Ca2+-ATPase inhibitor thapsigargin, and the chemotherapeutic agent, cisplatin. We first determined if these emetogens could regulate the phosphorylation level of GSK-3 in the brainstem emetic loci of least shrews and then investigated whether AR-A014418 and SB216763 could protect against the evoked emesis. Phospho-GSK-3α/β Ser21/9 levels in the brainstem and the enteric nerves of jejunum in the small intestine were upregulated following intraperitoneal (i.p.) administration of all the tested emetogens. Furthermore, administration of AR-A014418 (2.5-20 mg/kg, i.p.) dose-dependently attenuated both the frequency and percentage of shrews vomiting in response to i.p. administration of 5-HT (5 mg/kg), 2-Methyl-5-HT (5 mg/kg), GR73632 (5 mg/kg), apomorphine (2 mg/kg), quinpirole (2 mg/kg), pilocarpine (2 mg/kg), McN-A-343 (2 mg/kg), FPL64176 (10 mg/kg), or thapsigargin (0.5 mg/kg). Relatively lower doses of SB216763 exerted antiemetic efficacy, but both inhibitors barely affected cisplatin (10 mg/kg)-induced vomiting. Collectively, these results support the notion that vomiting is accompanied by a downregulation of GSK-3 activity and pharmacological inhibition of GSK-3 protects against pharmacologically evoked vomiting.
Collapse
Affiliation(s)
- W Zhong
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 East Second Street, Pomona, CA, 91766, USA
| | - N A Darmani
- Department of Basic Medical Sciences, College of Osteopathic Medicine of the Pacific, Western University of Health Sciences, 309 East Second Street, Pomona, CA, 91766, USA.
| |
Collapse
|
122
|
Paudel YN, Angelopoulou E, Jones NC, O’Brien TJ, Kwan P, Piperi C, Othman I, Shaikh MF. Tau Related Pathways as a Connecting Link between Epilepsy and Alzheimer's Disease. ACS Chem Neurosci 2019; 10:4199-4212. [PMID: 31532186 DOI: 10.1021/acschemneuro.9b00460] [Citation(s) in RCA: 31] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
Emerging findings point toward an important interconnection between epilepsy and Alzheimer's disease (AD) pathogenesis. Patients with epilepsy (PWE) commonly exhibit cognitive impairment similar to AD patients, who in turn are at a higher risk of developing epilepsy compared to age-matched controls. To date, no disease-modifying treatment strategy is available for either epilepsy or AD, reflecting an immediate need for exploring common molecular targets, which can delineate a possible mechanistic link between epilepsy and AD. This review attempts to disentangle the interconnectivity between epilepsy and AD pathogenesis via the crucial contribution of Tau protein. Tau protein is a microtubule-associated protein (MAP) that has been implicated in the pathophysiology of both epilepsy and AD. Hyperphosphorylation of Tau contributes to the different forms of human epilepsy and inhibition of the same exerted seizure inhibitions and altered disease progression in a range of animal models. Moreover, Tau-protein-mediated therapy has demonstrated promising outcomes in experimental models of AD. In this review, we discuss how Tau-related mechanisms might present a link between the cause of seizures in epilepsy and cognitive disruption in AD. Untangling this interconnection might be instrumental in designing novel therapies that can minimize epileptic seizures and cognitive deficits in patients with epilepsy and AD.
Collapse
Affiliation(s)
- Yam Nath Paudel
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 46150, Malaysia
| | - Efthalia Angelopoulou
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens 10679, Greece
| | - Nigel C. Jones
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne 3800, Australia
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, Victoria 3010, Australia
| | - Terence J. O’Brien
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne 3800, Australia
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, Victoria 3010, Australia
| | - Patrick Kwan
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne 3800, Australia
- Department of Medicine (Royal Melbourne Hospital), The University of Melbourne, Royal Parade, Parkville, Victoria 3010, Australia
| | - Christina Piperi
- Department of Biological Chemistry, Medical School, National and Kapodistrian University of Athens, Athens 10679, Greece
| | - Iekhsan Othman
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 46150, Malaysia
| | - Mohd. Farooq Shaikh
- Neuropharmacology Research Laboratory, Jeffrey Cheah School of Medicine and Health Sciences, Monash University Malaysia, Bandar Sunway, Selangor 46150, Malaysia
- Department of Neuroscience, Central Clinical School, Monash University, The Alfred Hospital, Melbourne 3800, Australia
| |
Collapse
|
123
|
Cauli O. Neuro-Psychiatric Disorders: From Diagnosis to Care. Diseases 2019; 7:diseases7030048. [PMID: 31284423 PMCID: PMC6787584 DOI: 10.3390/diseases7030048] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2019] [Accepted: 07/04/2019] [Indexed: 11/16/2022] Open
Abstract
Neuro-psychiatric disorders are an important cause of poor quality of life, disability, and premature mortality [...]
Collapse
Affiliation(s)
- Omar Cauli
- Department of Nursing, University of Valencia, 46010 Valencia, Spain.
| |
Collapse
|
124
|
Komatsu H, Fukuchi M, Habata Y. Potential Utility of Biased GPCR Signaling for Treatment of Psychiatric Disorders. Int J Mol Sci 2019; 20:E3207. [PMID: 31261897 PMCID: PMC6651563 DOI: 10.3390/ijms20133207] [Citation(s) in RCA: 18] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2019] [Revised: 06/24/2019] [Accepted: 06/25/2019] [Indexed: 12/12/2022] Open
Abstract
Tremendous advances have been made recently in the identification of genes and signaling pathways associated with the risks for psychiatric disorders such as schizophrenia and bipolar disorder. However, there has been a marked reduction in the pipeline for the development of new psychiatric drugs worldwide, mainly due to the complex causes that underlie these disorders. G-protein coupled receptors (GPCRs) are the most common targets of antipsychotics such as quetiapine and aripiprazole, and play pivotal roles in controlling brain function by regulating multiple downstream signaling pathways. Progress in our understanding of GPCR signaling has opened new possibilities for selective drug development. A key finding has been provided by the concept of biased ligands, which modulate some, but not all, of a given receptor's downstream signaling pathways. Application of this concept raises the possibility that the biased ligands can provide therapeutically desirable outcomes with fewer side effects. Instead, this application will require a detailed understanding of the mode of action of antipsychotics that drive distinct pharmacologies. We review our current understanding of the mechanistic bases for multiple signaling modes by antipsychotics and the potential of the biased modulators to treat mental disorders.
Collapse
Affiliation(s)
- Hidetoshi Komatsu
- Medical Affairs, Kyowa Pharmaceutical Industry Co., Ltd. (A Lupin Group Company), Osaka 530-0005, Japan.
- Department of Biological Science, Graduate School of Science, Nagoya University, Nagoya City 464-8602, Japan.
| | - Mamoru Fukuchi
- Laboratory of Molecular Neuroscience, Faculty of Pharmacy, Takasaki University of Health and Welfare, Gunma 370-0033, Japan
| | - Yugo Habata
- Department of Food & Nutrition, Yamanashi Gakuin Junior College, Kofu 400-8575, Japan
| |
Collapse
|