1
|
Mishra SK, Mishra V. Saroglitazar Enhances Memory Functions and Adult Neurogenesis via Up-Regulation of Wnt/β Catenin Signaling in the Rat Model of Dementia. ACS Chem Neurosci 2024; 15:3449-3458. [PMID: 39265183 DOI: 10.1021/acschemneuro.4c00167] [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] [Indexed: 09/14/2024] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) have emerged as a promising target for the treatment of various neurodegenerative disorders. Studies have shown that both PPAR α & γ individually modulate various pathophysiological events like neuroinflammation and insulin resistance, which are known to variedly affect neurogenesis. Our study aimed to evaluate the effect of saroglitazar (SGZR), a dual PPAR agonist, on adult neurogenesis and spatial learning and memory, in intracerebroventricular streptozotocin (ICV STZ)-induced dementia in rats. We have found that SGZR at the dose of 4 mg/kg per oral showed significant improvement in learning and memory compared to ICV STZ-treated rats. A substantial increase in neurogenesis was observed in the subventricular zone (SVZ) and the dentate gyrus (DG), as indicated by an increase in the number of 5-bromo-2'-deoxyuridine (BrdU)+ cells, BrdU+ nestin+ cells, and doublecortin (DCX)+cells. Treatment with SGZR also decreased the active form of glycogen synthase kinase 3β (GSK3β) and hence enhanced the nuclear translocation of the β-catenin. Enhanced expression of Wnt transcription factors and target genes indicates that the up-regulation of Wnt signaling might be involved in the observed increase in neurogenesis. Hence, it can be concluded that the SGZR enhances memory functions and adult neurogenesis via the upregulation of Wnt β-catenin signaling in ICV STZ-treated rats.
Collapse
Affiliation(s)
- Sandeep Kumar Mishra
- Division of Pharmacology, CSIR-Central Drug Research Institute, Lucknow, Uttar Pradesh 226031, India
- Faculty of Pharmacy, Kalinga University, Raipur, Chhattisgarh 492101, India
| | - Vaibhav Mishra
- Amity Institute of Microbial Technology, Amity University, Noida, Uttar Pradesh 201313, India
| |
Collapse
|
2
|
Carniglia L, Turati J, Saba J, López Couselo F, Romero AC, Caruso C, Durand D, Lasaga M. Melanocortin-receptor 4 activation modulates proliferation and differentiation of rat postnatal hippocampal neural precursor cells. Neuropharmacology 2024; 257:110058. [PMID: 38960135 DOI: 10.1016/j.neuropharm.2024.110058] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/08/2024] [Revised: 06/28/2024] [Accepted: 06/30/2024] [Indexed: 07/05/2024]
Abstract
Postnatal hippocampal neurogenesis is essential for learning and memory. Hippocampal neural precursor cells (NPCs) can be induced to proliferate and differentiate into either glial cells or dentate granule cells. Notably, hippocampal neurogenesis decreases dramatically with age, partly due to a reduction in the NPC pool and a decrease in their proliferative activity. Alpha-melanocyte-stimulating hormone (α-MSH) improves learning, memory, neuronal survival and plasticity. Here, we used postnatally-isolated hippocampal NPCs from Wistar rat pups (male and female combined) to determine the role of the melanocortin analog [Nle4, D-Phe7]-α-MSH (NDP-MSH) in proliferation and fate acquisition of NPCs. Incubation of growth-factor deprived NPCs with 10 nM NDP-MSH for 6 days increased the proportion of Ki-67- and 5-bromo-2'-deoxyuridine (BrdU)-positive cells, compared to the control group, and these effects were blocked by the MC4R antagonist JKC-363. NDP-MSH also increased the proportion of glial fibrillar acidic protein (GFAP)/Ki-67, GFAP/sex-determining region Y-box2 (SOX2) and neuroepithelial stem cell protein (NESTIN)/Ki-67-double positive cells (type-1 and type-2 precursors). Finally, NDP-MSH induced peroxisome proliferator-activated receptor (PPAR)-γ protein expression, and co-incubation with the PPAR-γ inhibitor GW9662 prevented the effect of NDP-MSH on NPC proliferation and differentiation. Our results indicate that in vitro activation of MC4R in growth-factor-deprived postnatal hippocampal NPCs induces proliferation and promotes the relative expansion of the type-1 and type-2 NPC pool through a PPAR-γ-dependent mechanism. These results shed new light on the mechanisms underlying the beneficial effects of melanocortins in hippocampal plasticity and provide evidence linking the MC4R and PPAR-γ pathways in modulation of hippocampal NPC proliferation and differentiation.
Collapse
Affiliation(s)
- Lila Carniglia
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Paraguay 2155, piso 10, CP 1121, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina.
| | - Juan Turati
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Paraguay 2155, piso 10, CP 1121, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Julieta Saba
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Paraguay 2155, piso 10, CP 1121, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Federico López Couselo
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Paraguay 2155, piso 10, CP 1121, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Ana Clara Romero
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Paraguay 2155, piso 10, CP 1121, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Carla Caruso
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Paraguay 2155, piso 10, CP 1121, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Daniela Durand
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Paraguay 2155, piso 10, CP 1121, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| | - Mercedes Lasaga
- Instituto de Investigaciones Biomédicas (INBIOMED, UBA-CONICET), Paraguay 2155, piso 10, CP 1121, Facultad de Medicina, Universidad de Buenos Aires, Ciudad Autónoma de Buenos Aires, Argentina
| |
Collapse
|
3
|
Queiroz LY, Kageyama R, Cimarosti HI. SUMOylation effects on neural stem cells self-renewal, differentiation, and survival. Neurosci Res 2024; 199:1-11. [PMID: 37742800 DOI: 10.1016/j.neures.2023.09.006] [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: 06/23/2023] [Revised: 09/14/2023] [Accepted: 09/20/2023] [Indexed: 09/26/2023]
Abstract
SUMO (small ubiquitin-like modifier) conjugation or SUMOylation, a post-translational modification, is a crucial regulator of protein function and cellular processes. In the context of neural stem cells (NSCs), SUMOylation has emerged as a key player, affecting their proliferation, differentiation, and survival. By modifying transcription factors, such as SOX1, SOX2, SOX3, SOX6, Bmi1, and Nanog, SUMOylation can either enhance or impair their transcriptional activity, thus impacting on NSCs self-renewal. Moreover, SUMOylation regulates neurogenesis and neuronal differentiation by modulating key proteins, such as Foxp1, Mecp2, MEF2A, and SOX10. SUMOylation is also crucial for the survival and proliferation of NSCs in both developing and adult brains. By regulating the activity of transcription factors, coactivators, and corepressors, SUMOylation acts as a molecular switch, inducing cofactor recruitment and function during development. Importantly, dysregulation of NSCs SUMOylation has been implicated in various disorders, including embryonic defects, ischemic cerebrovascular disease, glioma, and the harmful effects of benzophenone-3 exposure. Here we review the main findings on SUMOylation-mediated regulation of NSCs self-renewal, differentiation and survival. Better understanding NSCs SUMOylation mechanisms and its functional consequences might provide new strategies to promote neuronal differentiation that could contribute for the development of novel therapies targeting neurodegenerative diseases.
Collapse
Affiliation(s)
- Letícia Yoshitome Queiroz
- Postgraduate Program in Pharmacology, Federal University of Santa Catarina (UFSC), Florianopolis, Brazil
| | - Ryoichiro Kageyama
- Graduate School of Medicine, Kyoto University, Kyoto, Japan; RIKEN Center for Brain Science, Wako, Japan
| | - Helena I Cimarosti
- Postgraduate Program in Pharmacology, Federal University of Santa Catarina (UFSC), Florianopolis, Brazil; Postgraduate Program in Neuroscience, UFSC, Florianopolis, Brazil.
| |
Collapse
|
4
|
Newell AJ, Patisaul HB. Developmental organophosphate flame retardant exposure disrupts adult hippocampal neurogenesis in Wistar rats. Neurotoxicology 2023; 99:104-114. [PMID: 37783313 PMCID: PMC10842265 DOI: 10.1016/j.neuro.2023.09.009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/19/2023] [Revised: 09/26/2023] [Accepted: 09/28/2023] [Indexed: 10/04/2023]
Abstract
Organophosphate flame retardant (OPFR) contamination is ubiquitous and bio-monitoring studies have shown that human exposure is widespread and may be unavoidable. OPFRs bear structural similarities to known neurotoxicants such as organophosphate insecticides and have been shown to have both endocrine disrupting and developmental neurotoxic effects. The perinatal period in rodents represents a critical period in the organization of the developing nervous system and insults during this time can impart profound changes on the trajectory of neural development and function, lasting into adulthood. Adult hippocampal neurogenesis (AHN) facilitates dentate gyrus function and broader hippocampal circuit activity in adults; however, the neurogenic potential of this process in adulthood is vulnerable to disruption by exogenous factors during early life. We sought to assess the impact of OPFRs on AHN in offspring of dams exposed during gestation and lactation. Results indicate that developmental OPFR exposure has significant, sex specific impacts on multiple markers of AHN in the dentate gyrus of rats. In males, OPFR exposure significantly reduced the number of neural progenitors the number of new/immature neurons and reduced dentate gyrus volume. In females, exposure increased the number of neural progenitors, decreased the number of new/immature neurons, but had no significant effect on dentate gyrus volume. These results further elucidate the developmental neurotoxic properties of OPFRs, emphasize the long-term impact of early life OPFR exposure on neural processes, and highlight the importance of including sex as a biological variable in neurotoxicology research.
Collapse
Affiliation(s)
- Andrew J Newell
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA.
| | - Heather B Patisaul
- Department of Biological Sciences, North Carolina State University, Raleigh, NC 27695, USA; Center for Human Health and the Environment, North Carolina State University, Raleigh, NC 27695, USA
| |
Collapse
|
5
|
Nakao-Ise Y, Narita T, Miyamoto S, Watanabe M, Tanaka T, Sowa Y, Iizumi Y, Masuda M, Fujii G, Hirai Y, Nakao T, Takakura H, Mutoh M. Induction of MYCN-amplified neuroblastoma differentiation through NMYC suppression using PPAR-γ antagonist. J Clin Biochem Nutr 2023; 73:191-197. [PMID: 37970556 PMCID: PMC10636585 DOI: 10.3164/jcbn.23-28] [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: 04/27/2023] [Accepted: 06/21/2023] [Indexed: 11/17/2023] Open
Abstract
Neuroblastomas are the most common extracranial solid tumors in children and have a unique feature of neuronal differentiation. Peroxisome proliferator-activated receptor (PPAR)-γ is reported to have neuroprotective effects in addition to having antitumor effects in various cancers. Thus, we aimed to clarify the role of PPAR-γ agonist and antagonist in malignant neuroblastomas, which also possess neuronal features. In MYCN-amplified neuroblastoma CHP212 cells, treatment with the PPAR-γ antagonist GW9662 induced growth inhibition in a dose-dependent manner. In addition, the PPAR-γ antagonist treatment changed cell morphology with increasing expression of the neuronal differentiation marker tubulin beta 3 (TUBB3) and induced G1 phase arrest and apoptosis in MYCN-amplified neuroblastoma. Notably, the PPAR-γ antagonist treatment significantly decreased expression of NMYC, B-cell lymphoma 2 (BCL2) and bromodomain-containing protein 4 (BRD4). It is implied that BRD4, NMYC, BCL2 suppression by the PPAR-γ antagonist resulted in cell growth inhibition, differentiation, and apoptosis induction. In our in vivo study, the PPAR-γ antagonist treatment induced CHP212 cells differentiation and resultant tumor growth inhibition. Our results provide a deeper understanding of the mechanisms of tumor cell differentiation and suggest that PPAR-γ antagonist is a new therapeutic and prevention option for neuroblastomas.
Collapse
Affiliation(s)
- Yukako Nakao-Ise
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takumi Narita
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Shingo Miyamoto
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Motoki Watanabe
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Takuji Tanaka
- Department of Diagnostic Pathology & Research Center of Diagnostic Pathology, Gifu Municipal Hospital, 7-1 Kashima-cho, Gifu 500-8513, Japan
| | - Yoshihiro Sowa
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Yosuke Iizumi
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Mitsuharu Masuda
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Gen Fujii
- Central Radioisotope Division, National Cancer Center Research Institute, 5-1-1 Tsukiji, Chuo-ku, Tokyo 104-0045, Japan
| | - Yasuko Hirai
- Department of Human Immunology and Nutrition Science, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| | - Toshimasa Nakao
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
- Organ Transplantation Center, National Center for Child Health and Development, 2-10-1 Okura, Setagaya-ku, Tokyo 157-8535, Japan
| | - Hideki Takakura
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
- Laboratory of Biopharmaceutics and Pharmacokinetics, Faculty of Pharmaceutical Sciences, Hiroshima International University, 5-1-1 Hirokoshingai, Kure city, Hiroshima 737-0112, Japan
| | - Michihiro Mutoh
- Department of Molecular-Targeting Prevention, Kyoto Prefectural University of Medicine, Kawaramachi-Hirokoji, Kamigyo-ku, Kyoto 602-8566, Japan
| |
Collapse
|
6
|
Rayner MLD, Kellaway SC, Kingston I, Guillemot-Legris O, Gregory H, Healy J, Phillips JB. Exploring the Nerve Regenerative Capacity of Compounds with Differing Affinity for PPARγ In Vitro and In Vivo. Cells 2022; 12:cells12010042. [PMID: 36611836 PMCID: PMC9818498 DOI: 10.3390/cells12010042] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/18/2022] [Revised: 12/09/2022] [Accepted: 12/10/2022] [Indexed: 12/24/2022] Open
Abstract
Damage to peripheral nerves can cause debilitating consequences for patients such as lifelong pain and disability. At present, no drug treatments are routinely given in the clinic following a peripheral nerve injury (PNI) to improve regeneration and remyelination of damaged nerves. Appropriately targeted therapeutic agents have the potential to be used at different stages following nerve damage, e.g., to maintain Schwann cell viability, induce and sustain a repair phenotype to support axonal growth, or promote remyelination. The development of therapies to promote nerve regeneration is currently of high interest to researchers, however, translation to the clinic of drug therapies for PNI is still lacking. Studying the effect of PPARγ agonists for treatment of peripheral nerve injures has demonstrated significant benefits. Ibuprofen, a non-steroidal anti-inflammatory drug (NSAID), has reproducibly demonstrated benefits in vitro and in vivo, suggested to be due to its agonist action on PPARγ. Other NSAIDs have demonstrated differing levels of PPARγ activation based upon their affinity. Therefore, it was of interest to determine whether affinity for PPARγ of selected drugs corresponded to an increase in regeneration. A 3D co-culture in vitro model identified some correlation between these two properties. However, when the drug treatments were screened in vivo, in a crush injury model in a rat sciatic nerve, the same correlation was not apparent. Further differences were observed between capacity to increase axon number and improvement in functional recovery. Despite there not being a clear correlation between affinity and size of effect on regeneration, all selected PPARγ agonists improved regeneration, providing a panel of compounds that could be explored for use in the treatment of PNI.
Collapse
Affiliation(s)
- Melissa L. D. Rayner
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
- Correspondence:
| | - Simon C. Kellaway
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - Isabel Kingston
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - Owein Guillemot-Legris
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - Holly Gregory
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - Jess Healy
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| | - James B. Phillips
- Department of Pharmacology, School of Pharmacy, University College London, London WC1N 1AX, UK
- Centre for Nerve Engineering, University College London, London WC1N 6BT, UK
| |
Collapse
|
7
|
Alhowail A, Alsikhan R, Alsaud M, Aldubayan M, Rabbani SI. Protective Effects of Pioglitazone on Cognitive Impairment and the Underlying Mechanisms: A Review of Literature. Drug Des Devel Ther 2022; 16:2919-2931. [PMID: 36068789 PMCID: PMC9441149 DOI: 10.2147/dddt.s367229] [Citation(s) in RCA: 14] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2022] [Accepted: 08/23/2022] [Indexed: 11/23/2022] Open
Affiliation(s)
- Ahmad Alhowail
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al Qassim, 52452, Kingdom of Saudi Arabia
- Correspondence: Ahmad Alhowail, Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al Qassim, 52452, Kingdom of Saudi Arabia, Tel +9665672025858, Email
| | - Rawan Alsikhan
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al Qassim, 52452, Kingdom of Saudi Arabia
- Department of Pharmacology and Toxicology, Unaizah College of Pharmacy, Qassim University, Unaizah, Al Qassim, 51911, Kingdom of Saudi Arabia
| | - May Alsaud
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al Qassim, 52452, Kingdom of Saudi Arabia
| | - Maha Aldubayan
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al Qassim, 52452, Kingdom of Saudi Arabia
| | - Syed Imam Rabbani
- Department of Pharmacology and Toxicology, College of Pharmacy, Qassim University, Buraydah, Al Qassim, 52452, Kingdom of Saudi Arabia
| |
Collapse
|
8
|
Zhao G, Hu J, Gao M, Zhu Y, Hong Y. Excessive selenium affects neural development and locomotor behavior of zebrafish embryos. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2022; 238:113611. [PMID: 35526456 DOI: 10.1016/j.ecoenv.2022.113611] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/26/2022] [Revised: 04/29/2022] [Accepted: 05/04/2022] [Indexed: 06/14/2023]
Abstract
Selenium is an essential micronutrient derived from daily diet to maintain the normal growth and development of vertebrates. Excessive selenium intake will induce cardiovascular toxicity, reproductive toxicity and neurotoxicity. However, there have been few studies of the toxic effects of selenium on neural development and locomotor behavior. In this study, newly fertilized zebrafish embryos were treated with selenium. As a result, selenium treatment at the concentration of 0.5 µM decreased the moving speed and distance and blunted the touch response of zebrafish embryos. TUNEL assay and immunofluorescence analysis revealed that selenium induced nervous system impairment including promoted cell apoptosis, proliferation and neuroinflammation, and decreased neurons in zebrafish embryos. RNA-seq and RT-PCR results indicated that selenium treatment significantly decreased the expression of the dopaminergic neuron, motor neuron, GABAergic neuron and neurotransmitter transport marker genes in zebrafish embryos. The expression of PPAR signaling pathway marker genes was significantly down-regulated in selenium-treated embryos. Two PPAR agonists (rosiglitazone and bezafibrate) and an anti-cancer drug (cisplatin) were tested for their effects to alleviate selenium-induced locomotor defects. Rosiglitazone and bezafibrate could restore the expression of some neural marker genes but could not fully rescue the selenium-induced locomotor behavior defects. The supplementation of cisplatin could restore the dysfunctional locomotor behavior and the abnormal expression of the PPAR and neural marker genes to almost the normal levels. In conclusion, the results of this study reveal that selenium-induced neural development and locomotor behavior defects are caused by multiple complex factors including PPAR signaling, and all the factors might be recovered by cisplatin through unknown mechanisms.
Collapse
Affiliation(s)
- Guang Zhao
- School of Life Science, Key Laboratory of Aquatic Resources and Utilization of Jiangxi, Nanchang University, Nanchang 330031, China
| | - Jun Hu
- School of Life Science, Key Laboratory of Aquatic Resources and Utilization of Jiangxi, Nanchang University, Nanchang 330031, China
| | - Meng Gao
- School of Life Science, Key Laboratory of Aquatic Resources and Utilization of Jiangxi, Nanchang University, Nanchang 330031, China
| | - Yuejie Zhu
- School of Life Science, Key Laboratory of Aquatic Resources and Utilization of Jiangxi, Nanchang University, Nanchang 330031, China
| | - Yijiang Hong
- School of Life Science, Key Laboratory of Aquatic Resources and Utilization of Jiangxi, Nanchang University, Nanchang 330031, China.
| |
Collapse
|
9
|
Ye X, Wu Y, Xu J, Liu H, Wang H, Li Q, Li Q, Xuan A. PPARβ mediates mangiferin-induced neuronal differentiation of neural stem cells through DNA demethylation. Pharmacol Res 2022; 179:106235. [PMID: 35472635 DOI: 10.1016/j.phrs.2022.106235] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2022] [Revised: 04/16/2022] [Accepted: 04/21/2022] [Indexed: 11/21/2022]
Abstract
Adult hippocampal neurogenesis (AHN) is heavily implicated in the pathogenesis of various neuropsychiatric disorders. The mangiferin (MGF), a bioactive compound of the mango, reportedly produces biological effects on a variety of neuropsychiatric disorders. However, the function and underlying mechanisms of MGF in regulating hippocampal neurogenesis remain unknown. Here we discovered that the transcriptome and methylome of MGF-induced neural stem cells (NSCs) are distinct from the control. RNA-seq analysis revealed that the diferentially expressed genes (DEGs) were signifcantly enriched in the PPARs. Furthermore, we found that MGF enhanced neuronal differentiation and proliferation of neural stem cells (NSCs) via PPARβ but not PPARα and PPARγ. The combination of WGBS and RNA-seq analysis showed that the expression of some neurogenesis genes was negatively correlated with the DNA methylation level generally. We further found that PPARβ increased demethylation of Mash1 promoter by modulating the expressions of active and passive DNA demethylation enzymes in MGF-treated NSCs. Importantly, genetic deficiency of PPARβ decreased hippocampal neurogenesis in the adult mice, whereas the defective neurogenesis was notably rescued by Mash1 overexpression. Our findings uncover a model that PPARβ-mediated DNA demethylation of Mash1 contributes to MGF-induced neuronal genesis, and advance the concept that targeting PPARβ-TET1/DNMT3a-Mash1 axis regulation of neurogenesis might serve as a novel neurotherapeutic strategy.
Collapse
Affiliation(s)
- Xiujuan Ye
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, School of Basic Medical Sciences of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, China
| | - Yuanfei Wu
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, School of Basic Medical Sciences of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, China
| | - Jiamin Xu
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, School of Basic Medical Sciences of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, China
| | - Hui Liu
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, School of Basic Medical Sciences of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, China
| | - Huan Wang
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, School of Basic Medical Sciences of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, China
| | - Qingfeng Li
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, School of Basic Medical Sciences of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, China
| | - Qingqing Li
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, School of Basic Medical Sciences of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, China
| | - Aiguo Xuan
- Institute of Neuroscience and the Second Affiliated Hospital of Guangzhou Medical University, School of Basic Medical Sciences of Guangzhou Medical University, Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Guangzhou 510260, China.
| |
Collapse
|
10
|
Durai P, Beeraka NM, Ramachandrappa HVP, Krishnan P, Gudur P, Raghavendra NM, Ravanappa PKB. Advances in PPARs Molecular Dynamics and Glitazones as a Repurposing Therapeutic Strategy through Mitochondrial Redox Dynamics against Neurodegeneration. Curr Neuropharmacol 2022; 20:893-915. [PMID: 34751120 PMCID: PMC9881103 DOI: 10.2174/1570159x19666211109141330] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2021] [Revised: 08/20/2021] [Accepted: 09/17/2021] [Indexed: 11/22/2022] Open
Abstract
Peroxisome proliferator-activated receptors (PPARs) activity has significant implications for the development of novel therapeutic modalities against neurodegenerative diseases. Although PPAR-α, PPAR-β/δ, and PPAR-γ nuclear receptor expressions are significantly reported in the brain, their implications in brain physiology and other neurodegenerative diseases still require extensive studies. PPAR signaling can modulate various cell signaling mechanisms involved in the cells contributing to on- and off-target actions selectively to promote therapeutic effects as well as the adverse effects of PPAR ligands. Both natural and synthetic ligands for the PPARα, PPARγ, and PPARβ/δ have been reported. PPARα (WY 14.643) and PPARγ agonists can confer neuroprotection by modulating mitochondrial dynamics through the redox system. The pharmacological effect of these agonists may deliver effective clinical responses by protecting vulnerable neurons from Aβ toxicity in Alzheimer's disease (AD) patients. Therefore, the current review delineated the ligands' interaction with 3D-PPARs to modulate neuroprotection, and also deciphered the efficacy of numerous drugs, viz. Aβ aggregation inhibitors, vaccines, and γ-secretase inhibitors against AD; this review elucidated the role of PPAR and their receptor isoforms in neural systems, and neurodegeneration in human beings. Further, we have substantially discussed the efficacy of PPREs as potent transcription factors in the brain, and the role of PPAR agonists in neurotransmission, PPAR gamma coactivator-1α (PGC-1α) and mitochondrial dynamics in neuroprotection during AD conditions. This review concludes with the statement that the development of novel PPARs agonists may benefit patients with neurodegeneration, mainly AD patients, which may help mitigate the pathophysiology of dementia, subsequently improving overall the patient's quality of life.
Collapse
Affiliation(s)
- Priya Durai
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru 570 015, India and JSS Academy of Higher Education & Research, Mysuru, Karnataka, India
| | - Narasimha M. Beeraka
- Center of Excellence in Regenerative Medicine and Molecular Biology (CEMR), Department of Biochemistry, JSS Medical College, JSS Academy of Higher Education & Research, Mysuru 570 015, Karnataka, India;,I.M. Sechenov First Moscow State Medical University (Sechenov University), Moscow 119146, Russia
| | - Hemanth Vikram Poola Ramachandrappa
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru 570 015, India and JSS Academy of Higher Education & Research, Mysuru, Karnataka, India
| | | | - Pranesh Gudur
- Swamy Vivekananda Yoga Anusandhana Samsthana Deemed University, Bengaluru 560 105, India
| | | | - Prashantha Kumar Bommenahally Ravanappa
- Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru 570 015, India and JSS Academy of Higher Education & Research, Mysuru, Karnataka, India;,Address correspondence to this author at the Department of Pharmaceutical Chemistry, JSS College of Pharmacy, Mysuru 570 015, India and JSS Academy of Higher Education & Research (JSS AHER), Mysuru, Karnataka, India; E-mail:
| |
Collapse
|
11
|
Laurent AJ, Bindslev N, Vukojević V, Terenius L. Iso-α-acids in Nonalcoholic and Alcoholic Beer Stimulate Growth of Neuron-like SH-SY5Y Cells and Neuroepithelial Stem Cells. ACS BIO & MED CHEM AU 2021; 1:11-20. [PMID: 37101982 PMCID: PMC10125168 DOI: 10.1021/acsbiomedchemau.1c00017] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
With the increasing popularity of nonalcoholic beer, the association between beer drinking and alcohol intake is lost. In the present study, we show that nonalcoholic beer can stimulate the expansion of neuron-like cell lines and neuroepithelial stem cells in culture, yielding an effect comparable to that of alcoholic beer. One ingredient in beer is hops, which is derived from the flower of hop plants. The female flower contains humulones, which are transformed into iso-α-acids during wort boiling and give beer its bitter taste. In this study, we tested the effects of these iso-α-acids and/or alcohol on the proliferation of neuron-like cells and neuroepithelial stem cells in culture. Iso-α-acids enhanced cell expansion, showing a bimodal dose-response curve with peaks around 2-30 nM and 2-5 μM, of which nanomolar concentrations are relevant in beer drinking. The more lipophilic trans-iso-α-acids, found to a greater extent in beer foam, are even more potent. Our results indicate that iso-α-acids, acting via peroxisome proliferator-activated receptors could be responsible for the observed effects. Altogether, our results indicate that nonalcoholic beer with ingredients such as iso-α-acids stimulate the proliferation of neuroepithelial stem cells.
Collapse
Affiliation(s)
- Agneta J. Laurent
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Niels Bindslev
- Department of Biomedical Sciences, Faculty of Health and Medical Sciences, University of Copenhagen, Blegdamsvej 3B, DK-2200 Copenhagen N, Denmark
| | - Vladana Vukojević
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| | - Lars Terenius
- Department of Clinical Neuroscience, Center for Molecular Medicine, Karolinska Institutet, SE-171 76 Stockholm, Sweden
| |
Collapse
|
12
|
PPAR Gamma and Viral Infections of the Brain. Int J Mol Sci 2021; 22:ijms22168876. [PMID: 34445581 PMCID: PMC8396218 DOI: 10.3390/ijms22168876] [Citation(s) in RCA: 16] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2021] [Revised: 08/04/2021] [Accepted: 08/06/2021] [Indexed: 12/29/2022] Open
Abstract
Peroxisome Proliferator-Activated Receptor gamma (PPARγ) is a master regulator of metabolism, adipogenesis, inflammation and cell cycle, and it has been extensively studied in the brain in relation to inflammation or neurodegeneration. Little is known however about its role in viral infections of the brain parenchyma, although they represent the most frequent cause of encephalitis and are a major threat for the developing brain. Specific to viral infections is the ability to subvert signaling pathways of the host cell to ensure virus replication and spreading, as deleterious as the consequences may be for the host. In this respect, the pleiotropic role of PPARγ makes it a critical target of infection. This review aims to provide an update on the role of PPARγ in viral infections of the brain. Recent studies have highlighted the involvement of PPARγ in brain or neural cells infected by immunodeficiency virus 1, Zika virus, or human cytomegalovirus. They have provided a better understanding on PPARγ functions in the infected brain, and revealed that it can be a double-edged sword with respect to inflammation, viral replication, or neuronogenesis. They unraveled new roles of PPARγ in health and disease and could possibly help designing new therapeutic strategies.
Collapse
|
13
|
Mannan A, Garg N, Singh TG, Kang HK. Peroxisome Proliferator-Activated Receptor-Gamma (PPAR-ɣ): Molecular Effects and Its Importance as a Novel Therapeutic Target for Cerebral Ischemic Injury. Neurochem Res 2021; 46:2800-2831. [PMID: 34282491 DOI: 10.1007/s11064-021-03402-1] [Citation(s) in RCA: 24] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2021] [Revised: 07/10/2021] [Accepted: 07/12/2021] [Indexed: 02/06/2023]
Abstract
Cerebral ischemic injury is a leading cause of death and long-term disability throughout the world. Peroxisome proliferator-activated receptor gamma (PPAR-ɣ) is a ligand-activated nuclear transcription factor that is a member of the PPAR family. PPAR-ɣ has been shown in several in vitro and in vivo models to prevent post-ischemic inflammation and neuronal damage by negatively controlling the expression of genes modulated by cerebral ischemic injury, indicating a neuroprotective effect during cerebral ischemic injury. A extensive literature review of PubMed, Medline, Bentham, Scopus, and EMBASE (Elsevier) databases was carried out to understand the nature of the extensive work done on the mechanistic role of Peroxisome proliferator activated receptor gamma and its modulation in Cerebral ischemic injury. PPAR-ɣ can interact with specific DNA response elements to control gene transcription and expression when triggered by its ligand. It regulates lipid metabolism, improves insulin sensitivity, modulates antitumor mechanisms, reduces oxidative stress, and inhibits inflammation. This review article provides insights on the current state of research into the neuroprotective effects of PPAR-ɣ in cerebral ischemic injury, as well as the cellular and molecular mechanisms by which these effects are modulated, such as inhibition of inflammation, reduction of oxidative stress, suppression of pro-apoptotic production, modulation of transcription factors, and restoration of injured tissue through neurogenesis and angiogenesis.
Collapse
Affiliation(s)
- Ashi Mannan
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | - Nikhil Garg
- Chitkara College of Pharmacy, Chitkara University, Punjab, India
| | | | - Harmeet Kaur Kang
- Chitkara School of Health Sciences, Chitkara University, Punjab, India
| |
Collapse
|
14
|
Peroxisome proliferator-activated receptor gamma: a novel therapeutic target for cognitive impairment and mood disorders that functions via the regulation of adult neurogenesis. Arch Pharm Res 2021; 44:553-563. [PMID: 34138417 DOI: 10.1007/s12272-021-01333-7] [Citation(s) in RCA: 17] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2021] [Accepted: 05/25/2021] [Indexed: 12/12/2022]
Abstract
The proliferation, differentiation, and migration of neural precursor cells occur not only during embryonic development but also within distinct regions of the adult brain through the process of adult neurogenesis. As neurogenesis can potentially regulate brain cognition and neuronal plasticity, the factors that enhance neurogenesis can be attractive therapeutic targets for improving cognitive function and regulating neurodegenerative and neuropsychiatric disorders, including affective and mood disorders. Peroxisome proliferator-activated receptors (PPARs) are a class of ligand-activated transcription factors belonging to the nuclear receptor superfamily. PPARγ is a target for insulin sensitizers and plays an essential role in regulating various metabolic processes, including adipogenesis and glucose homeostasis. Interestingly, evidence demonstrates the role of PPARγ activation in regulating neurogenesis. The pharmacological activation of PPARγ using specific ligands increases the proliferation and differentiation of neural stem cells in specific brain regions, including the hippocampus, and prevents neurodegeneration and improves cognition and anxiety/depression-like behaviors in animal models. We summarize here recent reports on the role of PPARγ in adult neurogenesis, as well as the mechanisms involved, and suggest that PPARγ can serve as a potential therapeutic target for neurological and/or neurodegenerative diseases.
Collapse
|
15
|
Saunders AM, Burns DK, Gottschalk WK. Reassessment of Pioglitazone for Alzheimer's Disease. Front Neurosci 2021; 15:666958. [PMID: 34220427 PMCID: PMC8243371 DOI: 10.3389/fnins.2021.666958] [Citation(s) in RCA: 31] [Impact Index Per Article: 10.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2021] [Accepted: 05/18/2021] [Indexed: 01/01/2023] Open
Abstract
Alzheimer's disease is a quintessential 'unmet medical need', accounting for ∼65% of progressive cognitive impairment among the elderly, and 700,000 deaths in the United States in 2020. In 2019, the cost of caring for Alzheimer's sufferers was $244B, not including the emotional and physical toll on caregivers. In spite of this dismal reality, no treatments are available that reduce the risk of developing AD or that offer prolonged mitiagation of its most devestating symptoms. This review summarizes key aspects of the biology and genetics of Alzheimer's disease, and we describe how pioglitazone improves many of the patholophysiological determinants of AD. We also summarize the results of pre-clinical experiments, longitudinal observational studies, and clinical trials. The results of animal testing suggest that pioglitazone can be corrective as well as protective, and that its efficacy is enhanced in a time- and dose-dependent manner, but the dose-effect relations are not monotonic or sigmoid. Longitudinal cohort studies suggests that it delays the onset of dementia in individuals with pre-existing type 2 diabetes mellitus, which small scale, unblinded pilot studies seem to confirm. However, the results of placebo-controlled, blinded clinical trials have not borne this out, and we discuss possible explanations for these discrepancies.
Collapse
Affiliation(s)
- Ann M. Saunders
- Zinfandel Pharmaceuticals, Inc., Chapel Hill, NC, United States
| | - Daniel K. Burns
- Zinfandel Pharmaceuticals, Inc., Chapel Hill, NC, United States
| | | |
Collapse
|
16
|
Basak S, Mallick R, Banerjee A, Pathak S, Duttaroy AK. Maternal Supply of Both Arachidonic and Docosahexaenoic Acids Is Required for Optimal Neurodevelopment. Nutrients 2021; 13:2061. [PMID: 34208549 PMCID: PMC8234848 DOI: 10.3390/nu13062061] [Citation(s) in RCA: 30] [Impact Index Per Article: 10.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 06/10/2021] [Accepted: 06/14/2021] [Indexed: 12/22/2022] Open
Abstract
During the last trimester of gestation and for the first 18 months after birth, both docosahexaenoic acid,22:6n-3 (DHA) and arachidonic acid,20:4n-6 (ARA) are preferentially deposited within the cerebral cortex at a rapid rate. Although the structural and functional roles of DHA in brain development are well investigated, similar roles of ARA are not well documented. The mode of action of these two fatty acids and their derivatives at different structural-functional roles and their levels in the gene expression and signaling pathways of the brain have been continuously emanating. In addition to DHA, the importance of ARA has been much discussed in recent years for fetal and postnatal brain development and the maternal supply of ARA and DHA. These fatty acids are also involved in various brain developmental processes; however, their mechanistic cross talks are not clearly known yet. This review describes the importance of ARA, in addition to DHA, in supporting the optimal brain development and growth and functional roles in the brain.
Collapse
Affiliation(s)
- Sanjay Basak
- Molecular Biology Division, ICMR-National Institute of Nutrition, Indian Council of Medical Research, Hyderabad 500 007, India;
| | - Rahul Mallick
- A.I. Virtanen Institute for Molecular Sciences, University of Eastern Finland, 70210 Kuopio, Finland;
| | - Antara Banerjee
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai 603 103, India; (A.B.); (S.P.)
| | - Surajit Pathak
- Department of Medical Biotechnology, Faculty of Allied Health Sciences, Chettinad Academy of Research and Education (CARE), Chettinad Hospital and Research Institute (CHRI), Kelambakkam, Chennai 603 103, India; (A.B.); (S.P.)
| | - Asim K. Duttaroy
- Department of Nutrition, Institute of Basic Medical Sciences, Faculty of Medicine, University of Oslo, 0317 Oslo, Norway
| |
Collapse
|
17
|
Sağır D. Dose-dependent effects of prenatal exposure of pioglitazone, the PPARγ agonist, on the hippocampus development and learning and memory performance of rat offspring. Toxicol Appl Pharmacol 2021; 421:115544. [PMID: 33894214 DOI: 10.1016/j.taap.2021.115544] [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: 02/08/2021] [Revised: 04/10/2021] [Accepted: 04/19/2021] [Indexed: 10/21/2022]
Abstract
It is known that pioglitazone, defined as a PPARγ agonist, has neuron-protective properties in nervous system disorders. The aim of this study is to investigate the effects of pioglitazone administration at different doses during prenatal period on the neurons, glial cells and learning-memory levels in the hippocampus of rat offspring. Pregnant rats were divided into three groups; Low-Dose Pioglitazone (LDP), High-Dose Pioglitazone (HDP) and control (C) (n = 3). Pregnant rats in the HDP and LDP groups were given pioglitazone at 30 mg/kg and 5 mg/kg doses, respectively, by gavage once a day during their pregnancy. No procedure was applied to the rats in the control group. Morris water tank test was applied to offspring obtained from postnatal 24th to 28th day. The offspring were sacrificed on the 29th postal day and their brain tissues removed. Stereological, histopathological and immunohistochemical techniques were used to analyze brain tissues. As a result of the analysis, it was observed that there were delays in learning and memory, the number of pyramidal neurons decreased, and the density of cells stained with glial fibrillar acidic protein (GFAP) positive increased in the HDP group compared to the other groups (p < 0.05). No significant difference was found between the LDP and control groups in terms of these parameters (p > 0.05). Our results showed that pioglitazone administered in the prenatal period had an effect on the hippocampus development and learning and memory performance of rats, depending on the dose.
Collapse
Affiliation(s)
- Dilek Sağır
- Faculty of Health Sciences, Sinop University, 57000 Sinop, Turkey.
| |
Collapse
|
18
|
Li Y, Ren T, Xu L, Wang Y, Yang B, Luo H, Zeng Z, Zhang Y, Du G, Zhu M, Zhou J. Propane-2-sulfonic acid octadec-9-enyl-amide, a novel peroxisome proliferator-activated receptors α and γ dual agonist, enhances hippocampal neurogenesis and neuroplasticity in rats with cerebral ischaemia. Neuroreport 2020; 30:1299-1306. [PMID: 31714482 DOI: 10.1097/wnr.0000000000001360] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Our previous studies showed that propane-2-sulfonic acid octadec-9-enyl-amide (N15), a novel peroxisome proliferator-activated receptors α and γ (PPARα/γ) dual agonist, protected against ischaemia-induced acute brain damage in mice and improved cognitive ability in the chronic phase of ischaemic stroke. It is well known that hippocampal neurogenesis is closely related to cognitive function. In the present study, we investigated the effect of N15 on hippocampal neurogenesis and neuroplasticity in a middle cerebral artery occlusion (MCAO) rat model. The middle cerebral artery of rats was blocked for 2 hours. Oral administration of 100 mg/kg N15 or vehicle was given once daily for days 2-13 after MCAO. The newly mature neurons were detected by staining. The expressions of synapse-related proteins were observed by qRT-PCR or western blotting. We found that N15-treated rats showed improved survival post-MCAO. In addition, N15 treatment markedly increased the newly mature neurons and enhanced the expression levels of growth-associated protein-43, synaptophysin, brain-derived neurotrophic factor and neurotrophin-3 in the hippocampus. Moreover, N15 promoted the activation of PPARα and PPARγ on day 7 and 14 after cerebral ischaemia. These results reveal that N15 may promote neurogenesis and neuroplasticity in MCAO rats through the activation of the PPARα/γ dual signal pathway.
Collapse
Affiliation(s)
- Ying Li
- Department of Pharmacy, Xiamen Medical College
| | - Tong Ren
- Key Laboratory of Chiral Drugs, School of Medicine, Xiamen University
| | - Lanxi Xu
- Key Laboratory of Chiral Drugs, School of Medicine, Xiamen University
| | - Ying Wang
- Department of Pharmacy, Xiamen Medical College
| | - Bingye Yang
- Department of Pharmacy, Xiamen Medical College
| | - Haohong Luo
- Department of Pharmacy, Xiamen Medical College
| | - Zhen Zeng
- Department of Pharmacy, Xiamen Medical College
| | - Yanan Zhang
- Department of Pharmacy, Xiamen Medical College
| | - Guicheng Du
- Department of Pharmacy, Xiamen Medical College
| | - Maoshu Zhu
- Xiang'an Branch, The First Affiliated Hospital of Xiamen University.,The Fifth Hospital of Xiamen
| | - Juan Zhou
- Department of Obstetrics and Gynecology, The First Affiliated Hospital of Xiamen University, Xiamen, Fujian, China
| |
Collapse
|
19
|
Chounchay S, Noctor SC, Chutabhakdikul N. Microglia enhances proliferation of neural progenitor cells in an in vitro model of hypoxic-ischemic injury. EXCLI JOURNAL 2020; 19:950-961. [PMID: 32788909 PMCID: PMC7415932 DOI: 10.17179/excli2020-2249] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 04/07/2020] [Accepted: 06/24/2020] [Indexed: 12/18/2022]
Abstract
Microglial cells are the primary immune cells in the central nervous system. In the mature brain, microglia perform functions that include eliminating pathogens and clearing dead/dying cells and cellular debris through phagocytosis. In the immature brain, microglia perform functions that include synapse development and the regulation of cell production through extensive contact with and phagocytosis of neural progenitor cells (NPCs). However, the functional role of microglia in the proliferation and differentiation of NPCs under hypoxic-ischemic (HI) injury is not clear. Here, we tested the hypothesis that microglia enhance NPCs proliferation following HI insult. Primary NPCs cultures were divided into four treatment groups: 1) normoxic NPCs (NN); 2) normoxic NPCs cocultured with microglia (NN+M); 3) hypoxic NPCs (HN); and 4) hypoxic NPCs cocultured with microglia (HN+M). Hypoxic-ischemic injury was induced by pretreatment of the cell cultures with 100 µM deferoxamine mesylate (DFO). NPCs treated with 100 µM DFO (HN groups) for 24 hours had significantly increased expression of hypoxia-inducible factor 1 alpha (HIF-1α), a marker of hypoxic cells. Cell number, protein expression, mitosis, and cell cycle phase were examined, and the data were compared between the four groups. We found that the number of cells expressing the NPCs marker Sox2 increased significantly in the HN+M group and that the number of PH3-positive cells increased in the HN+M group; flow cytometry analysis showed a significant increase in the percentage of cells in the G2/M phase in the HN+M group. In summary, these results support the concept that microglia enhance the survival of NPCs under HI injury by increasing NPCs proliferation, survival, and differentiation. These results further suggest that microglia may induce neuroprotective effects after hypoxic injury that can be explored to develop novel therapeutic strategies for the treatment of HI injury in the immature brain.
Collapse
Affiliation(s)
- Supanee Chounchay
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhonpathom, 73170, Thailand.,Faculty of Physical Therapy, Huachiew Chalermprakiet University, Samut Prakan, 10540, Thailand
| | - Stephen C Noctor
- Department of Psychiatry and Behavioral Sciences, University of California, Davis, Sacramento, CA, 95817, USA.,MIND Institute, University of California, Davis, Sacramento, CA, 95817, USA
| | - Nuanchan Chutabhakdikul
- Research Center for Neuroscience, Institute of Molecular Biosciences, Mahidol University, Salaya, Nakhonpathom, 73170, Thailand
| |
Collapse
|
20
|
Elastin-derived peptide VGVAPG affects the proliferation of mouse cortical astrocytes with the involvement of aryl hydrocarbon receptor (Ahr), peroxisome proliferator-activated receptor gamma (Pparγ), and elastin-binding protein (EBP). Cytokine 2019; 126:154930. [PMID: 31760184 DOI: 10.1016/j.cyto.2019.154930] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2019] [Revised: 11/13/2019] [Accepted: 11/14/2019] [Indexed: 12/17/2022]
Abstract
During aging and ischemic and hemorrhagic stroke, elastin molecules are degraded and elastin-derived peptides are released into the brain microenvironment. Val-Gly-Val-Ala-Pro-Gly (VGVAPG) is a repeating hexapeptide in the elastin molecule. It is well documented that the peptide sequence binds with high affinity to elastin-binding protein (EBP) located on the cell surface, thereby transducing a molecular signal into the cell. The aim of our study was to investigate whether EBP, aryl hydrocarbon receptor (Ahr), and peroxisome proliferator-activated receptor gamma (Pparγ) are involved in VGVAPG-stimulated proliferation. Primary astrocytes were maintained in DMEM/F12 medium without phenol red, supplemented with 10 or 1% charcoal/dextran-treated fetal bovine serum (FBS). The cells were exposed to increasing concentrations of VGVAPG peptide, and resazurin reduction was measured. In addition, Glb1, Pparγ, and Ahr genes were silenced. After 48 h of exposure to 10 nM and 1 µM of VGVAPG peptide, the level of estradiol (E2) and the expression of Ki67 and S100B proteins were measured. The results showed that at a wide range of concentrations, VGVAPG peptide increased the metabolism of astrocytes depending on the concentration of FBS. After silencing of Glb1, Pparγ, and Ahr genes, VGVAPG peptide did not affect the cell metabolism which suggests the involvement of all the mentioned receptors in its mechanism of action. Interestingly, in the low-FBS medium, the silencing of Glb1 gene did not result in complete inhibition of VGVAPG-stimulated proliferation. On the other hand, in the medium with 10% FBS VGVAPG increased Ki67 expression after Pparγ silencing, whereas in the medium with 1% FBS VGVAPG decreased Ki67 expression. Following the application of Ahr siRNA, VGVAPG peptide decreased the production of E2 and increased the expression of Ki67 and S100B proteins.
Collapse
|
21
|
Docosahexaenoic acid,22:6n-3: Its roles in the structure and function of the brain. Int J Dev Neurosci 2019; 79:21-31. [PMID: 31629800 DOI: 10.1016/j.ijdevneu.2019.10.004] [Citation(s) in RCA: 58] [Impact Index Per Article: 11.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2019] [Revised: 10/10/2019] [Accepted: 10/11/2019] [Indexed: 12/12/2022] Open
Abstract
Docosahexaenoic acid,22:6n-3 (DHA) and its metabolites are vital for the structure and functional brain development of the fetus and infants, and also for maintenance of healthy brain function of adults. DHA is thought to be an essential nutrient required throughout the life cycle for the maintenance of overall brain health. The mode of actions of DHA and its derivatives at both cellular and molecular levels in the brain are emerging. DHA is the major prevalent fatty acid in the brain membrane. The brain maintains its fatty acid levels mainly via the uptake of plasma free fatty acids. Therefore, circulating plasma DHA is significantly related to cognitive abilities during ageing and is inversely associated with cognitive decline. The signaling pathways of DHA and its metabolites are involved in neurogenesis, antinociceptive effects, anti-apoptotic effect, synaptic plasticity, Ca2+ homeostasis in brain diseases, and the functioning of nigrostriatal activities. Mechanisms of action of DHA metabolites on various processes in the brain are not yet well known. Epidemiological studies support a link between low habitual intake of DHA and a higher risk of brain disorders. A diet characterized by higher intakes of foods containing high in n-3 fatty acids, and/or lower intake of n-6 fatty acids was strongly associated with a lower Alzheimer's Disease and other brain disorders. Supplementation of DHA improves some behaviors associated with attention deficit hyperactivity disorder, bipolar disorder, schizophrenia, and impulsive behavior, as well as cognition. Nevertheless, the outcomes of trials with DHA supplementation have been controversial. Many intervention studies with DHA have shown an apparent benefit in brain function. However, clinical trials are needed for definitive conclusions. Dietary deficiency of n-3 fatty acids during fetal development in utero and the postnatal state has detrimental effects on cognitive abilities. Further research in humans is required to assess a variety of clinical outcomes, including quality of life and mental status, by supplementation of DHA.
Collapse
|
22
|
Du F, Yuelling L, Lee EH, Wang Y, Liao S, Cheng Y, Zhang L, Zheng C, Peri S, Cai KQ, Ng JMY, Curran T, Li P, Yang ZJ. Leukotriene Synthesis Is Critical for Medulloblastoma Progression. Clin Cancer Res 2019; 25:6475-6486. [PMID: 31300449 DOI: 10.1158/1078-0432.ccr-18-3549] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2018] [Revised: 04/18/2019] [Accepted: 07/02/2019] [Indexed: 12/22/2022]
Abstract
PURPOSE Here, we examined the role of leukotrienes, well-known inflammatory mediators, in the tumorigenesis of hedgehog pathway-associated medulloblastoma, and tested the efficacies of antagonists of leukotriene biosynthesis in medulloblastoma treatment.Experimental Design: We examined the leukotriene levels in medulloblastoma cells by ELISA. We next tested whether leukotriene synthesis in medulloblastoma cells relied on activation of hedgehog pathway, or the presence of hedgehog ligand secreted by astrocytes. We then investigated whether leukotriene mediated hedgehog-induced Nestin expression in tumor cells. The functions of leukotriene in tumor cell proliferation and tumor growth in medulloblastoma were determined through knocking down 5-lipoxygenase (a critical enzyme for leukotriene synthesis) by shRNAs, or using 5-lipoxygenase-deficient mice. Finally, the efficacies of antagonists of leukotriene synthesis in medulloblastoma treatment were tested in vivo and in vitro. RESULTS Leukotriene was significantly upregulated in medulloblastoma cells. Increased leukotriene synthesis relied on hedgehog ligand secreted by astrocytes, a major component of medulloblastoma microenvironment. Leukotriene stimulated tumor cells to express Nestin, a cytoskeletal protein essential for medulloblastoma growth. Genetic blockage of leukotriene synthesis dramatically suppressed medulloblastoma cell proliferation and tumor growth in vivo. Pharmaceutical inhibition of leukotriene synthesis markedly repressed medulloblastoma cell proliferation, but had no effect on proliferation of normal neuronal progenitors. Moreover, antagonists of leukotriene synthesis exhibited promising tumor inhibitory efficacies on drug-resistant medulloblastoma. CONCLUSIONS Our findings reveal a novel signaling pathway that is critical for medulloblastoma cell proliferation and tumor progression, and that leukotriene biosynthesis represents a promising therapeutic target for medulloblastoma treatment.
Collapse
Affiliation(s)
- Fang Du
- Laboratory of Molecular Neuropathology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China.,Cancer Biology Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Larra Yuelling
- Cancer Biology Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Eric H Lee
- Cancer Biology Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Yuan Wang
- Laboratory of Molecular Neuropathology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Shengyou Liao
- Laboratory of Molecular Neuropathology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Yan Cheng
- Laboratory of Molecular Neuropathology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China.,Cancer Biology Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Li Zhang
- Laboratory of Molecular Neuropathology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Chaonan Zheng
- Laboratory of Molecular Neuropathology, College of Pharmaceutical Sciences, Soochow University, Suzhou, China
| | - Suraj Peri
- Biostatistics and Bioinformatics Research Facility, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Kathy Q Cai
- Cancer Biology Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania
| | - Jessica M Y Ng
- Children's Research Institute, Children's Mercy Kansas City, Kansas City, Missouri
| | - Tom Curran
- Children's Research Institute, Children's Mercy Kansas City, Kansas City, Missouri
| | - Peng Li
- Department of Pharmacognosy and Traditional Chinese Pharmacology, College of Pharmacy, Army Medical University, Chongqing, China
| | - Zeng-Jie Yang
- Cancer Biology Program, Fox Chase Cancer Center, Temple University Health System, Philadelphia, Pennsylvania.
| |
Collapse
|
23
|
Szychowski KA, Rombel-Bryzek A, Dołhańczuk-Śródka A, Gmiński J. Antiproliferative Effect of Elastin-Derived Peptide VGVAPG on SH-SY5Y Neuroblastoma Cells. Neurotox Res 2019; 36:503-514. [PMID: 31161598 PMCID: PMC6745029 DOI: 10.1007/s12640-019-00040-y] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2019] [Revised: 03/29/2019] [Accepted: 04/01/2019] [Indexed: 02/06/2023]
Abstract
Throughout the lifetime of humans, the amount of stem cells and the rate of cell proliferation continue to decrease. Reactive oxygen species (ROS) are one among the many factors that promote stem cell aging. Both a decrease in the level of stem cells and increase in ROS production can lead to the development of different neurodegenerative diseases. This study was conducted to determine how the VGVAPG peptide, liberated from elastin during the aging process and under pathological conditions, affects ROS production and activities of antioxidant enzymes in undifferentiated, proliferating SH-SY5Y cells. SH-SY5Y cells were maintained in Dulbecco's modified Eagle's medium/nutrient mixture F-12 supplemented with 10% heat-inactivated fetal bovine serum (FBS). After treating the SH-SY5Y cells with VGVAPG peptide, we measured ROS production; cell metabolism, proliferation, and expression; and activities of superoxide dismutase (SOD), glutathione peroxidase (GPx), and catalase (CAT). We demonstrated that the VGVAPG peptide increases GPx expression and activity, whereas it decreases CAT expression in SH-SY5Y cells. Silencing of the GLB1 gene prevents changes in GPx activity. Despite the fact that the VGVAPG peptide increases GPx expression, it increases the ROS level. Moreover, the VGVAPG peptide decreases SH-SY5Y proliferation, which is prevented by the ROS scavenger N-acetyl-L-cysteine. Our data suggest that ROS production and decreased proliferation of SH-SY5Y cells are the results of excitotoxicity meditated through close unrecognized molecular pathways. More research is needed to elucidate the unknown mechanism of action of VGVAPG peptide in the nervous system.
Collapse
Affiliation(s)
- Konrad A Szychowski
- Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medicine, University of Opole, Oleska 48, 45-052, Opole, Poland.
| | - Agnieszka Rombel-Bryzek
- Department of Clinical Biochemistry and Laboratory Diagnostics, Institute of Medicine, University of Opole, Oleska 48, 45-052, Opole, Poland
| | | | - Jan Gmiński
- Department of Public Health, Dietetics and Lifestyle Disorders, Faculty of Medicine, University of Information Technology and Management in Rzeszow, Sucharskiego 2, 35-225, Rzeszow, Poland
| |
Collapse
|
24
|
Lee DW, Kim MS, Lim YH, Lee N, Hong YC. Prenatal and postnatal exposure to di-(2-ethylhexyl) phthalate and neurodevelopmental outcomes: A systematic review and meta-analysis. ENVIRONMENTAL RESEARCH 2018; 167:558-566. [PMID: 30145432 DOI: 10.1016/j.envres.2018.08.023] [Citation(s) in RCA: 50] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/23/2018] [Revised: 07/09/2018] [Accepted: 08/16/2018] [Indexed: 06/08/2023]
Abstract
BACKGROUND Di-(2-ethylhexyl) phthalate (DEHP), the most widely used phthalate, has recently been associated with neurodevelopmental disturbances in children. However, the risk is yet to be quantified. Therefore, a systematic review and meta-analysis focusing on the association between exposure to DEHP and neurodevelopmental outcomes is necessary, with particular attention to study design (longitudinal vs. cross-sectional). METHODS We performed a comprehensive literature search for associations between exposure to DEHP and neurodevelopmental outcomes. Among 106 published studies found in public databases, eight longitudinal studies and two cross-sectional studies were included in the meta-analysis. RESULTS We observed a statistically significant association between the concentrations of DEHP metabolites and the neurodevelopment outcomes of children among cross-sectional results, and found significant association between DEHP exposure measured in prenatal period and the psychomotor development outcomes measured later in childhood. CONCLUSIONS To our knowledge, this is the first meta-analysis of studies investigating the association between DEHP exposure and neurodevelopment in children. A need exists for more researches and a precautionary policy for potential health hazard of DEHP, the most commonly used phthalate, to promote healthier neurodevelopment in children.
Collapse
Affiliation(s)
- Dong-Wook Lee
- Department of Preventive Medicine, College of Medicine, Seoul National University, 103 Daehangno, Jongno-gu, Seoul 110-799, Republic of Korea
| | - Min-Seok Kim
- Department of Preventive Medicine, College of Medicine, Seoul National University, 103 Daehangno, Jongno-gu, Seoul 110-799, Republic of Korea
| | - Youn-Hee Lim
- Environmental Health Centre, College of Medicine, Seoul National University, Seoul, Republic of Korea; Institute of Environmental Medicine, Medical Research Centre, Seoul National University, Seoul, Republic of Korea
| | - Nami Lee
- Department of Psychiatry, College of Medicine, Seoul National University, Seoul, Republic of Korea
| | - Yun-Chul Hong
- Department of Preventive Medicine, College of Medicine, Seoul National University, 103 Daehangno, Jongno-gu, Seoul 110-799, Republic of Korea; Environmental Health Centre, College of Medicine, Seoul National University, Seoul, Republic of Korea; Institute of Environmental Medicine, Medical Research Centre, Seoul National University, Seoul, Republic of Korea.
| |
Collapse
|
25
|
Menzel-Severing J, Zenkel M, Polisetti N, Sock E, Wegner M, Kruse FE, Schlötzer-Schrehardt U. Transcription factor profiling identifies Sox9 as regulator of proliferation and differentiation in corneal epithelial stem/progenitor cells. Sci Rep 2018; 8:10268. [PMID: 29980721 PMCID: PMC6035181 DOI: 10.1038/s41598-018-28596-3] [Citation(s) in RCA: 30] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2018] [Accepted: 06/26/2018] [Indexed: 02/08/2023] Open
Abstract
Understanding transcription factor (TF) regulation of limbal epithelial stem/progenitor cells (LEPCs) may aid in using non-ocular cells to regenerate the corneal surface. This study aimed to identify and characterize TF genes expressed specifically in LEPCs isolated from human donor eyes by laser capture microdissection. Using a profiling approach, preferential limbal expression was found for SoxE and SoxF genes, particularly for Sox9, which showed predominantly cytoplasmic localization in basal LEPCs and nuclear localization in suprabasal and corneal epithelial cells, indicating nucleocytoplasmic translocation and activation during LEPC proliferation and differentiation. Increased nuclear localization of Sox9 was also observed in activated LEPCs following clonal expansion and corneal epithelial wound healing. Knockdown of SOX9 expression in cultured LEPCs by RNAi led to reduced expression of progenitor cell markers, e.g. keratin 15, and increased expression of differentiation markers, e.g. keratin 3. Furthermore, SOX9 silencing significantly suppressed the proliferative capacity of LEPCs and reduced levels of glycogen synthase kinase 3 beta (GSK-3ß), a negative regulator of Wnt/ß-catenin signaling. Sox9 expression, in turn, was significantly suppressed by treatment of LEPCs with exogenous GSK-3ß inhibitors and enhanced by small molecule inhibitors of Wnt signaling. Our results suggest that Sox9 and Wnt/ß-catenin signaling cooperate in mutually repressive interactions to achieve a balance between quiescence, proliferation and differentiation of LEPCs in the limbal niche. Future molecular dissection of Sox9-Wnt interaction and mechanisms of nucleocytoplasmic shuttling of Sox9 may aid in improving the regenerative potential of LEPCs and the reprogramming of non-ocular cells for corneal surface regeneration.
Collapse
Affiliation(s)
- Johannes Menzel-Severing
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Matthias Zenkel
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Naresh Polisetti
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Elisabeth Sock
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Michael Wegner
- Institut für Biochemie, Emil-Fischer-Zentrum, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | - Friedrich E Kruse
- Department of Ophthalmology, Friedrich-Alexander-Universität Erlangen-Nürnberg, Erlangen, Germany
| | | |
Collapse
|
26
|
D'Angelo M, Antonosante A, Castelli V, Catanesi M, Moorthy N, Iannotta D, Cimini A, Benedetti E. PPARs and Energy Metabolism Adaptation during Neurogenesis and Neuronal Maturation. Int J Mol Sci 2018; 19:ijms19071869. [PMID: 29949869 PMCID: PMC6073366 DOI: 10.3390/ijms19071869] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/07/2018] [Revised: 06/20/2018] [Accepted: 06/24/2018] [Indexed: 11/20/2022] Open
Abstract
Peroxisome proliferator activated receptors (PPARs) are a class of ligand-activated transcription factors, belonging to the superfamily of receptors for steroid and thyroid hormones, retinoids, and vitamin D. PPARs control the expression of several genes connected with carbohydrate and lipid metabolism, and it has been demonstrated that PPARs play important roles in determining neural stem cell (NSC) fate. Lipogenesis and aerobic glycolysis support the rapid proliferation during neurogenesis, and specific roles for PPARs in the control of different phases of neurogenesis have been demonstrated. Understanding the changes in metabolism during neuronal differentiation is important in the context of stem cell research, neurodegenerative diseases, and regenerative medicine. In this review, we will discuss pivotal evidence that supports the role of PPARs in energy metabolism alterations during neuronal maturation and neurodegenerative disorders.
Collapse
Affiliation(s)
- Michele D'Angelo
- 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.
| | - 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.
| | - NandhaKumar Moorthy
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy.
| | - Dalila Iannotta
- 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.
| | - Elisabetta Benedetti
- Department of Life, Health and Environmental Sciences, University of L'Aquila, 67100 L'Aquila, Italy.
| |
Collapse
|
27
|
Combined Rosiglitazone and Forskolin Have Neuroprotective Effects in SD Rats after Spinal Cord Injury. PPAR Res 2018; 2018:3897478. [PMID: 30034460 PMCID: PMC6032969 DOI: 10.1155/2018/3897478] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Revised: 04/22/2018] [Accepted: 05/08/2018] [Indexed: 02/05/2023] Open
Abstract
The peroxisome proliferator-activated receptor gamma (PPAR-γ) agonist rosiglitazone inhibits NF-κB expression and endogenous neural stem cell differentiation into neurons and reduces the inflammatory cascade after spinal cord injury (SCI). The aim of this study was to explore the mechanisms underlying rosiglitazone-mediated neuroprotective effects and regulation of the balance between the inflammatory cascade and generation of endogenous spinal cord neurons by using a spinal cord-derived neural stem cell culture system as well as SD rat SCI model. Activation of PPAR-γ could promote neural stem cell proliferation and inhibit PKA expression and neuronal formation in vitro. In the SD rat SCI model, the rosiglitazone + forskolin group showed better locomotor recovery compared to the rosiglitazone and forskolin groups. MAP2 expression was higher in the rosiglitazone + forskolin group than in the rosiglitazone group, NF-κB expression was lower in the rosiglitazone + forskolin group than in the forskolin group, and NeuN expression was higher in the rosiglitazone + forskolin group than in the forskolin group. PPAR-γ activation likely inhibits NF-κB, thereby reducing the inflammatory cascade, and PKA activation likely promotes neuronal cell regeneration.
Collapse
|
28
|
Oral pioglitazone ameliorates fructose-induced peripheral insulin resistance and hippocampal gliosis but not restores inhibited hippocampal adult neurogenesis. Biochim Biophys Acta Mol Basis Dis 2018; 1864:274-285. [DOI: 10.1016/j.bbadis.2017.10.017] [Citation(s) in RCA: 13] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2017] [Revised: 09/13/2017] [Accepted: 10/12/2017] [Indexed: 12/12/2022]
|
29
|
Gkikas D, Tsampoula M, Politis PK. Nuclear receptors in neural stem/progenitor cell homeostasis. Cell Mol Life Sci 2017; 74:4097-4120. [PMID: 28638936 PMCID: PMC11107725 DOI: 10.1007/s00018-017-2571-4] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2017] [Revised: 06/06/2017] [Accepted: 06/13/2017] [Indexed: 12/13/2022]
Abstract
In the central nervous system, embryonic and adult neural stem/progenitor cells (NSCs) generate the enormous variety and huge numbers of neuronal and glial cells that provide structural and functional support in the brain and spinal cord. Over the last decades, nuclear receptors and their natural ligands have emerged as critical regulators of NSC homeostasis during embryonic development and adult life. Furthermore, substantial progress has been achieved towards elucidating the molecular mechanisms of nuclear receptors action in proliferative and differentiation capacities of NSCs. Aberrant expression or function of nuclear receptors in NSCs also contributes to the pathogenesis of various nervous system diseases. Here, we review recent advances in our understanding of the regulatory roles of steroid, non-steroid, and orphan nuclear receptors in NSC fate decisions. These studies establish nuclear receptors as key therapeutic targets in brain diseases.
Collapse
Affiliation(s)
- Dimitrios Gkikas
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Str, 115 27, Athens, Greece
| | - Matina Tsampoula
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Str, 115 27, Athens, Greece
| | - Panagiotis K Politis
- Center for Basic Research, Biomedical Research Foundation of the Academy of Athens, 4 Soranou Efesiou Str, 115 27, Athens, Greece.
| |
Collapse
|
30
|
O'Léime CS, Cryan JF, Nolan YM. Nuclear deterrents: Intrinsic regulators of IL-1β-induced effects on hippocampal neurogenesis. Brain Behav Immun 2017; 66:394-412. [PMID: 28751020 DOI: 10.1016/j.bbi.2017.07.153] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/03/2017] [Revised: 07/15/2017] [Accepted: 07/23/2017] [Indexed: 12/11/2022] Open
Abstract
Hippocampal neurogenesis, the process by which new neurons are born and develop into the host circuitry, begins during embryonic development and persists throughout adulthood. Over the last decade considerable insights have been made into the role of hippocampal neurogenesis in cognitive function and the cellular mechanisms behind this process. Additionally, an increasing amount of evidence exists on the impact of environmental factors, such as stress and neuroinflammation on hippocampal neurogenesis and subsequent impairments in cognition. Elevated expression of the pro-inflammatory cytokine interleukin-1β (IL-1β) in the hippocampus is established as a significant contributor to the neuronal demise evident in many neurological and psychiatric disorders and is now known to negatively regulate hippocampal neurogenesis. In order to prevent the deleterious effects of IL-1β on neurogenesis it is necessary to identify signalling pathways and regulators of neurogenesis within neural progenitor cells that can interact with IL-1β. Nuclear receptors are ligand regulated transcription factors that are involved in modulating a large number of cellular processes including neurogenesis. In this review we focus on the signalling mechanisms of specific nuclear receptors involved in regulating neurogenesis (glucocorticoid receptors, peroxisome proliferator activated receptors, estrogen receptors, and nuclear receptor subfamily 2 group E member 1 (NR2E1 or TLX)). We propose that these nuclear receptors could be targeted to inhibit neuroinflammatory signalling pathways associated with IL-1β. We discuss their potential to be therapeutic targets for neuroinflammatory disorders affecting hippocampal neurogenesis and associated cognitive function.
Collapse
Affiliation(s)
- Ciarán S O'Léime
- Department of Anatomy and Neuroscience, University College Cork, Ireland
| | - John F Cryan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland
| | - Yvonne M Nolan
- Department of Anatomy and Neuroscience, University College Cork, Ireland; APC Microbiome Institute, University College Cork, Ireland.
| |
Collapse
|
31
|
Chiang MC, Cheng YC, Nicol CJ, Lin CH. The neuroprotective role of rosiglitazone in advanced glycation end product treated human neural stem cells is PPARgamma-dependent. Int J Biochem Cell Biol 2017; 92:121-133. [DOI: 10.1016/j.biocel.2017.09.020] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2017] [Revised: 09/08/2017] [Accepted: 09/26/2017] [Indexed: 12/11/2022]
|
32
|
Cai W, Yang T, Liu H, Han L, Zhang K, Hu X, Zhang X, Yin KJ, Gao Y, Bennett MVL, Leak RK, Chen J. Peroxisome proliferator-activated receptor γ (PPARγ): A master gatekeeper in CNS injury and repair. Prog Neurobiol 2017; 163-164:27-58. [PMID: 29032144 DOI: 10.1016/j.pneurobio.2017.10.002] [Citation(s) in RCA: 158] [Impact Index Per Article: 22.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2017] [Revised: 10/06/2017] [Accepted: 10/08/2017] [Indexed: 01/06/2023]
Abstract
Peroxisome proliferator-activated receptor γ (PPARγ) is a widely expressed ligand-modulated transcription factor that governs the expression of genes involved in inflammation, redox equilibrium, trophic factor production, insulin sensitivity, and the metabolism of lipids and glucose. Synthetic PPARγ agonists (e.g. thiazolidinediones) are used to treat Type II diabetes and have the potential to limit the risk of developing brain injuries such as stroke by mitigating the influence of comorbidities. If brain injury develops, PPARγ serves as a master gatekeeper of cytoprotective stress responses, improving the chances of cellular survival and recovery of homeostatic equilibrium. In the acute injury phase, PPARγ directly restricts tissue damage by inhibiting the NFκB pathway to mitigate inflammation and stimulating the Nrf2/ARE axis to neutralize oxidative stress. During the chronic phase of acute brain injuries, PPARγ activation in injured cells culminates in the repair of gray and white matter, preservation of the blood-brain barrier, reconstruction of the neurovascular unit, resolution of inflammation, and long-term functional recovery. Thus, PPARγ lies at the apex of cell fate decisions and exerts profound effects on the chronic progression of acute injury conditions. Here, we review the therapeutic potential of PPARγ in stroke and brain trauma and highlight the novel role of PPARγ in long-term tissue repair. We describe its structure and function and identify the genes that it targets. PPARγ regulation of inflammation, metabolism, cell fate (proliferation/differentiation/maturation/survival), and many other processes also has relevance to other neurological diseases. Therefore, PPARγ is an attractive target for therapies against a number of progressive neurological disorders.
Collapse
Affiliation(s)
- Wei Cai
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Tuo Yang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Huan Liu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Lijuan Han
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Kai Zhang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Xiaoming Hu
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai 200032, China; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh PA, USA
| | - Xuejing Zhang
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Ke-Jie Yin
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA
| | - Yanqin Gao
- State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai 200032, China
| | - Michael V L Bennett
- Dominick P. Purpura Department of Neuroscience, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | - Rehana K Leak
- Division of Pharmaceutical Sciences, School of Pharmacy, Duquesne University, Pittsburgh, PA 15282, USA.
| | - Jun Chen
- Pittsburgh Institute of Brain Disorders & Recovery and Department of Neurology, University of Pittsburgh, Pittsburgh, PA 15213, USA; State Key Laboratory of Medical Neurobiology and Institutes of Brain Science, Fudan University, Shanghai 200032, China; Geriatric Research, Education and Clinical Center, Veterans Affairs Pittsburgh Health Care System, Pittsburgh PA, USA.
| |
Collapse
|
33
|
Roles of Peroxisome Proliferator-Activated Receptor Gamma on Brain and Peripheral Inflammation. Cell Mol Neurobiol 2017; 38:121-132. [PMID: 28975471 DOI: 10.1007/s10571-017-0554-5] [Citation(s) in RCA: 232] [Impact Index Per Article: 33.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2017] [Accepted: 09/23/2017] [Indexed: 02/08/2023]
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) has been implicated in the pathology of numerous diseases involving diabetes, stroke, cancer, or obesity. It is expressed in diverse cell types, including vessels, immune and glial cells, and neurons. PPARγ plays crucial roles in the regulation of cellular differentiation, lipid metabolism, or glucose homeostasis. PPARγ ligands also exert effects on attenuating degenerative processes in the brain, as well as in peripheral systems, and it has been associated with the control of anti-inflammatory mechanisms, oxidative stress, neuronal death, neurogenesis, differentiation, and angiogenesis. This review will highlight key advances in the understanding of the PPARγ-related mechanisms responsible for neuroprotection after brain injuries, both ischemia and traumatic brain injury, and it will also cover the natural and synthetic agonist for PPARγ, angiotensin receptor blockers, and PPARγ antagonists, used in experimental and clinical research. A better understanding of the pleiotropic mechanisms and applications of these drugs to improve the recovery and to repair the acute and chronic induced neuroinflammation after brain injuries will pave the way for more effective therapeutic strategies after brain deficits.
Collapse
|
34
|
Pleniceanu O, Shukrun R, Omer D, Vax E, Kanter I, Dziedzic K, Pode-Shakked N, Mark-Daniei M, Pri-Chen S, Gnatek Y, Alfandary H, Varda-Bloom N, Bar-Lev DD, Bollag N, Shtainfeld R, Armon L, Urbach A, Kalisky T, Nagler A, Harari-Steinberg O, Arbiser JL, Dekel B. Peroxisome proliferator-activated receptor gamma (PPARγ) is central to the initiation and propagation of human angiomyolipoma, suggesting its potential as a therapeutic target. EMBO Mol Med 2017; 9:508-530. [PMID: 28275008 PMCID: PMC5376758 DOI: 10.15252/emmm.201506111] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Angiomyolipoma (AML), the most common benign renal tumor, can result in severe morbidity from hemorrhage and renal failure. While mTORC1 activation is involved in its growth, mTORC1 inhibitors fail to eradicate AML, highlighting the need for new therapies. Moreover, the identity of the AML cell of origin is obscure. AML research, however, is hampered by the lack of in vivo models. Here, we establish a human AML‐xenograft (Xn) model in mice, recapitulating AML at the histological and molecular levels. Microarray analysis demonstrated tumor growth in vivo to involve robust PPARG‐pathway activation. Similarly, immunostaining revealed strong PPARG expression in human AML specimens. Accordingly, we demonstrate that while PPARG agonism accelerates AML growth, PPARG antagonism is inhibitory, strongly suppressing AML proliferation and tumor‐initiating capacity, via a TGFB‐mediated inhibition of PDGFB and CTGF. Finally, we show striking similarity between AML cell lines and mesenchymal stem cells (MSCs) in terms of antigen and gene expression and differentiation potential. Altogether, we establish the first in vivo human AML model, which provides evidence that AML may originate in a PPARG‐activated renal MSC lineage that is skewed toward adipocytes and smooth muscle and away from osteoblasts, and uncover PPARG as a regulator of AML growth, which could serve as an attractive therapeutic target.
Collapse
Affiliation(s)
- Oren Pleniceanu
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Hematology and Cord Blood Bank, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Racheli Shukrun
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Dorit Omer
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Einav Vax
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Itamar Kanter
- Faculty of Engineering, Institute of Nanotechnology, Bar-Ilan University, Ramat Gan, Israel
| | - Klaudyna Dziedzic
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Naomi Pode-Shakked
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Michal Mark-Daniei
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Sara Pri-Chen
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Yehudit Gnatek
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Hadas Alfandary
- Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel.,Institute of Nephrology, Schneider Children's Medical Center of Israel, Petah Tikva, Israel
| | - Nira Varda-Bloom
- Division of Hematology and Cord Blood Bank, Sheba Medical Center, Ramat Gan, Israel
| | - Dekel D Bar-Lev
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Naomi Bollag
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Rachel Shtainfeld
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Leah Armon
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Achia Urbach
- The Mina and Everard Goodman Faculty of Life Sciences, Bar-Ilan University, Ramat Gan, Israel
| | - Tomer Kalisky
- Faculty of Engineering, Institute of Nanotechnology, Bar-Ilan University, Ramat Gan, Israel
| | - Arnon Nagler
- Division of Hematology and Cord Blood Bank, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | - Orit Harari-Steinberg
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel
| | - Jack L Arbiser
- Department of Dermatology, Emory University School of Medicine, Atlanta, GA, USA.,Winship Cancer Institute, Atlanta Veterans Administration Hospital, Atlanta, GA, USA
| | - Benjamin Dekel
- Pediatric Stem Cell Research Institute, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel .,Division of Pediatric Nephrology, Edmond and Lily Safra Children's Hospital, Sheba Medical Center, Ramat Gan, Israel.,Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| |
Collapse
|
35
|
Lysophosphatidic acid signaling regulates the KLF9-PPARγ axis in human induced pluripotent stem cell-derived neurons. Biochem Biophys Res Commun 2017; 491:223-227. [DOI: 10.1016/j.bbrc.2017.07.082] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2017] [Accepted: 07/13/2017] [Indexed: 11/18/2022]
|
36
|
Zhang Y, Li X, Fang S, Zhu Z, Yao M, Ying L, Zhu L, Ma Z, Wang W. Peroxisome proliferator-activated receptor γ agonist suppresses mast cell maturation and induces apoptosis. Mol Med Rep 2017; 16:1793-1800. [PMID: 28656266 PMCID: PMC5562075 DOI: 10.3892/mmr.2017.6802] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2016] [Accepted: 06/08/2017] [Indexed: 01/15/2023] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPAR γ), is important in the immunoregulation of the allergic response. Mast cells are the most important inflammatory cells in immediate hypersensitivity and allergic diseases. However, there is limited information regarding the effects of PPAR γ on mast cell maturation. In the present study, mouse bone marrow-derived mast cells (BMMCs) were cultured in interleukin (IL)-3 and stem cell factor (SCF), in the presence or absence of the PPAR γ agonist, pioglitazone (PIO). The expression levels of the tyrosine kinase receptor CD117 and the high affinity IgE receptor FcεRI α, were assessed by flow cytometry, cell viability was assessed by Alamar-Blue assay and histamine release was determined by measuring the activity of β-hexosaminidase. IL-3 and SCF are required for the development of mast cells in vitro. PIO dose-dependently inhibited the expression of CD117 and FcεRI α, and the maturation of BMMCs. Treatment with PIO additionally inhibited the formation of granules and reduced the expression of β-hexosaminidase. In addition, reverse transcription-polymerase chain reaction analysis revealed that BMMCs treated with PIO expressed a lower level of mast cell protease (MCP)-6 mRNA and PIO treatment enhanced the level of PPAR γ mRNA. Furthermore, PIO induced mast cell progenitor apoptosis. PPAR γ agonists may maintain mast cell homeostasis by inhibiting maturation of their precursors. The inhibitory effects of PPAR γ agonists include suppression of the activation of mast cells and a decrease in mast cell function in the inflammatory response. Therefore, PPAR γ agonists may serve as effective anti-inflammatory reagents in the treatment of allergic reactions.
Collapse
Affiliation(s)
- Yu Zhang
- Department of Otolaryngology‑Head and Neck Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Xinqian Li
- Department of Otolaryngology‑Head and Neck Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Shengjian Fang
- Department of Otolaryngology‑Head and Neck Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Zhenghua Zhu
- Department of Otolaryngology‑Head and Neck Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Min Yao
- Department of Otolaryngology‑Head and Neck Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Liyun Ying
- Department of Otolaryngology‑Head and Neck Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Liwei Zhu
- Department of Otolaryngology‑Head and Neck Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Zhaoxin Ma
- Department of Otolaryngology‑Head and Neck Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| | - Weihua Wang
- Department of Otolaryngology‑Head and Neck Surgery, Shanghai East Hospital, Tongji University School of Medicine, Shanghai 200120, P.R. China
| |
Collapse
|
37
|
Physiological and Pathological Roles of 15-Deoxy-Δ12,14-Prostaglandin J2 in the Central Nervous System and Neurological Diseases. Mol Neurobiol 2017; 55:2227-2248. [DOI: 10.1007/s12035-017-0435-4] [Citation(s) in RCA: 17] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2016] [Accepted: 02/03/2017] [Indexed: 12/29/2022]
|
38
|
John S, Mishra R. mRNA Transcriptomics of Galectins Unveils Heterogeneous Organization in Mouse and Human Brain. Front Mol Neurosci 2016; 9:139. [PMID: 28018170 PMCID: PMC5159438 DOI: 10.3389/fnmol.2016.00139] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2016] [Accepted: 11/23/2016] [Indexed: 12/22/2022] Open
Abstract
Background: Galectins, a family of non-classically secreted, β-galactoside binding proteins is involved in several brain disorders; however, no systematic knowledge on the normal neuroanatomical distribution and functions of galectins exits. Hence, the major purpose of this study was to understand spatial distribution and predict functions of galectins in brain and also compare the degree of conservation vs. divergence between mouse and human species. The latter objective was required to determine the relevance and appropriateness of studying galectins in mouse brain which may ultimately enable us to extrapolate the findings to human brain physiology and pathologies. Results: In order to fill this crucial gap in our understanding of brain galectins, we analyzed the in situ hybridization and microarray data of adult mouse and human brain respectively, from the Allen Brain Atlas, to resolve each galectin-subtype’s spatial distribution across brain distinct cytoarchitecture. Next, transcription factors (TFs) that may regulate galectins were identified using TRANSFAC software and the list obtained was further curated to sort TFs on their confirmed transcript expression in the adult brain. Galectin-TF cluster analysis, gene-ontology annotations and co-expression networks were then extrapolated to predict distinct functional relevance of each galectin in the neuronal processes. Data shows that galectins have highly heterogeneous expression within and across brain sub-structures and are predicted to be the crucial targets of brain enriched TFs. Lgals9 had maximal spatial distribution across mouse brain with inferred predominant roles in neurogenesis while LGALS1 was ubiquitously expressed in human. Limbic region associated with learning, memory and emotions and substantia nigra associated with motor movements showed strikingly high expression of LGALS1 and LGALS8 in human vs. mouse brain. The overall expression profile of galectin-8 was most preserved across both these species, however, galectin-9 showed maximal preservation only in the cerebral cortex. Conclusion: It is for the first time that a comprehensive description of galectins’ mRNA expression profile in brain is presented. Results suggests that spatial transcriptome changes in galectins may contribute to differential brain functions and evolution across species that highlights galectins as novel signatures of brain heterogeneity and functions, which if disturbed, can promote several brain disorders.
Collapse
Affiliation(s)
- Sebastian John
- Disease Biology Program, Department of Neurobiology and Genetics, Rajiv Gandhi Centre for Biotechnology Thiruvananthapuram, India
| | - Rashmi Mishra
- Disease Biology Program, Department of Neurobiology and Genetics, Rajiv Gandhi Centre for Biotechnology Thiruvananthapuram, India
| |
Collapse
|
39
|
Di Giacomo E, Benedetti E, Cristiano L, Antonosante A, d'Angelo M, Fidoamore A, Barone D, Moreno S, Ippoliti R, Cerù MP, Giordano A, Cimini A. Roles of PPAR transcription factors in the energetic metabolic switch occurring during adult neurogenesis. Cell Cycle 2016; 16:59-72. [PMID: 27860527 DOI: 10.1080/15384101.2016.1252881] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/20/2022] Open
Abstract
PPARs are a class of ligand-activated transcription factors belonging to the superfamily of receptors for steroid and thyroid hormones, retinoids and vitamin D that control the expression of a large number of genes involved in lipid and carbohydrate metabolism and in the regulation of cell proliferation, differentiation and death. The role of PPARs in the CNS has been primarily associated with lipid and glucose metabolism; however, these receptors are also implicated in neural cell differentiation and death, as well as neuronal maturation. Although it has been demonstrated that PPARs play important roles in determining NSCs fate, less is known about their function in regulating NSCs metabolism during differentiation. In order to identify the metabolic events, controlled by PPARs, occurring during neuronal precursor differentiation, the glucose and lipid metabolism was followed in a recognized model of neuronal differentiation in vitro, the SH-SY5Y neuroblastoma cell line. Moreover, PPARs distribution were also followed in situ in adult mouse brains. The concept of adult neurogenesis becomes relevant especially in view of those disorders in which a loss of neurons is described, such as Alzheimer disease, Parkinson disease, brain injuries and other neurological disorders. Elucidating the crucial steps in energetic metabolism and the involvement of PPARγ in NSC neuronal fate (lineage) may be useful for the future design of preventive and/or therapeutic interventions.
Collapse
Affiliation(s)
- E Di Giacomo
- a Department of Life , Health and Environmental Sciences, University of L'Aquila , L'Aquila , Italy
| | - E Benedetti
- a Department of Life , Health and Environmental Sciences, University of L'Aquila , L'Aquila , Italy
| | - L Cristiano
- a Department of Life , Health and Environmental Sciences, University of L'Aquila , L'Aquila , Italy
| | - A Antonosante
- a Department of Life , Health and Environmental Sciences, University of L'Aquila , L'Aquila , Italy
| | - M d'Angelo
- a Department of Life , Health and Environmental Sciences, University of L'Aquila , L'Aquila , Italy
| | - A Fidoamore
- a Department of Life , Health and Environmental Sciences, University of L'Aquila , L'Aquila , Italy
| | - D Barone
- b Oncology Research Center of Mercogliano (CROM), Istituto Nazionale per lo Studio e la Cura dei Tumori "Fondazione Giovanni Pascale," IRCCS , Naples , Italy
| | - S Moreno
- c Department of Science-LIME , University Roma Tre , Rome , Italy
| | - R Ippoliti
- a Department of Life , Health and Environmental Sciences, University of L'Aquila , L'Aquila , Italy
| | - M P Cerù
- a Department of Life , Health and Environmental Sciences, University of L'Aquila , L'Aquila , Italy
| | - A Giordano
- d Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University , Philadelphia , PA , USA.,e Department of Medicine , Surgery and Neuroscience, University of Siena , Siena , Italy
| | - A Cimini
- a Department of Life , Health and Environmental Sciences, University of L'Aquila , L'Aquila , Italy.,d Sbarro Institute for Cancer Research and Molecular Medicine and Center for Biotechnology, Temple University , Philadelphia , PA , USA.,f National Institute for Nuclear Physics (INFN), Gran Sasso National Laboratory (LNGS) , Assergi , Italy
| |
Collapse
|
40
|
Chavanas S. Peroxisome proliferator-activated receptor γ (PPARγ) activation: A key determinant of neuropathogeny during congenital infection by cytomegalovirus. NEUROGENESIS 2016; 3:e1231654. [PMID: 27844024 DOI: 10.1080/23262133.2016.1231654] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Received: 06/08/2016] [Revised: 08/26/2016] [Accepted: 08/29/2016] [Indexed: 12/25/2022]
Abstract
Congenital infection by human cytomegalovirus (HCMV) might result in permanent neurological sequelae, including sensorineural deafness, cerebral palsies or devastating neurodevelopmental abnormalities. We recently disclosed that Peroxisome Proliferator-Activated Receptor gamma (PPARγ), a transcription factor of the nuclear receptor superfamily, is a key determinant of HCMV pathogenesis in developing brain. Using neural stem cells from human embryonic stem cells, we showed that HCMV infection strongly increases levels and activity of PPARγ in NSCs. Further in vitro experiments showed that PPARγ activity inhibits the neuronogenic differentiation of NSCs into neurons. Consistently, increased PPARγ expression was found in brain section of fetuses infected by HCMV, but not in uninfected controls. In this commentary, we summarize and discuss our findings and the new insights they provide on the neuropathogenesis of HCMV congenital infection.
Collapse
Affiliation(s)
- Stéphane Chavanas
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, Toulouse, France; CNRS UMR 5282 Toulouse, France; Université Paul Sabatier, Toulouse, France
| |
Collapse
|
41
|
PPAR Gamma in Neuroblastoma: The Translational Perspectives of Hypoglycemic Drugs. PPAR Res 2016; 2016:3038164. [PMID: 27799938 PMCID: PMC5069360 DOI: 10.1155/2016/3038164] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2016] [Accepted: 09/14/2016] [Indexed: 12/15/2022] Open
Abstract
Neuroblastoma (NB) is the most common and aggressive pediatric cancer, characterized by a remarkable phenotypic diversity and high malignancy. The heterogeneous clinical behavior, ranging from spontaneous remission to fatal metastatic disease, is attributable to NB biology and genetics. Despite major advances in therapies, NB is still associated with a high morbidity and mortality. Thus, novel diagnostic, prognostic, and therapeutic approaches are required, mainly to improve treatment outcomes of high-risk NB patients. Among neuroepithelial cancers, NB is the most studied tumor as far as PPAR ligands are concerned. PPAR ligands are endowed with antitumoral effects, mainly acting on cancer stem cells, and constitute a possible add-on therapy to antiblastic drugs, in particular for NB with unfavourable prognosis. While discussing clinical background, this review will provide a synopsis of the major studies about PPAR expression in NB, focusing on the potential beneficial effects of hypoglycemic drugs, thiazolidinediones and metformin, to reduce the occurrence of relapses as well as tumor regrowth in NB patients.
Collapse
|
42
|
Chang GQ, Karatayev O, Lukatskaya O, Leibowitz SF. Prenatal fat exposure and hypothalamic PPAR β/δ: Possible relationship to increased neurogenesis of orexigenic peptide neurons. Peptides 2016; 79:16-26. [PMID: 27002387 PMCID: PMC4872302 DOI: 10.1016/j.peptides.2016.03.007] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Revised: 03/17/2016] [Accepted: 03/18/2016] [Indexed: 01/12/2023]
Abstract
Gestational exposure to a fat-rich diet, while elevating maternal circulating fatty acids, increases in the offspring's hypothalamus and amygdala the proliferation and density of neurons that express neuropeptides known to stimulate consummatory behavior. To understand the relationship between these phenomena, this study examined in the brain of postnatal offspring (day 15) the effect of prenatal fat exposure on the transcription factor, peroxisome proliferator-activated receptor (PPAR) β/δ, which is sensitive to fatty acids, and the relationship of PPAR β/δ to the orexigenic neuropeptides, orexin, melanin-concentrating hormone, and enkephalin. Prenatal exposure to a fat-rich diet compared to low-fat chow increased the density of cells immunoreactive for PPAR β/δ in the hypothalamic paraventricular nucleus (PVN), perifornical lateral hypothalamus (PFLH), and central nucleus of the amygdala (CeA), but not the hypothalamic arcuate nucleus or basolateral amygdaloid nucleus. It also increased co-labeling of PPAR β/δ with the cell proliferation marker, BrdU, or neuronal marker, NeuN, and the triple labeling of PPAR β/δ with BrdU plus NeuN, indicating an increase in proliferation and density of new PPAR β/δ neurons. Prenatal fat exposure stimulated the double-labeling of PPAR β/δ with orexin or melanin-concentrating hormone in the PFLH and enkephalin in the PVN and CeA and also triple-labeling of PPAR β/δ with BrdU and these neuropeptides, indicating that dietary fat increases the genesis of PPAR β/δ neurons that produce these peptides. These findings demonstrate a close anatomical relationship between PPAR β/δ and the increased proliferation and density of peptide-expressing neurons in the hypothalamus and amygdala of fat-exposed offspring.
Collapse
Affiliation(s)
- G-Q Chang
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY, USA
| | - O Karatayev
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY, USA
| | - O Lukatskaya
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY, USA
| | - S F Leibowitz
- Laboratory of Behavioral Neurobiology, The Rockefeller University, New York, NY, USA.
| |
Collapse
|
43
|
Rolland M, Li X, Sellier Y, Martin H, Perez-Berezo T, Rauwel B, Benchoua A, Bessières B, Aziza J, Cenac N, Luo M, Casper C, Peschanski M, Gonzalez-Dunia D, Leruez-Ville M, Davrinche C, Chavanas S. PPARγ Is Activated during Congenital Cytomegalovirus Infection and Inhibits Neuronogenesis from Human Neural Stem Cells. PLoS Pathog 2016; 12:e1005547. [PMID: 27078877 PMCID: PMC4831785 DOI: 10.1371/journal.ppat.1005547] [Citation(s) in RCA: 32] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2015] [Accepted: 03/12/2016] [Indexed: 11/25/2022] Open
Abstract
Congenital infection by human cytomegalovirus (HCMV) is a leading cause of permanent sequelae of the central nervous system, including sensorineural deafness, cerebral palsies or devastating neurodevelopmental abnormalities (0.1% of all births). To gain insight on the impact of HCMV on neuronal development, we used both neural stem cells from human embryonic stem cells (NSC) and brain sections from infected fetuses and investigated the outcomes of infection on Peroxisome Proliferator-Activated Receptor gamma (PPARγ), a transcription factor critical in the developing brain. We observed that HCMV infection dramatically impaired the rate of neuronogenesis and strongly increased PPARγ levels and activity. Consistent with these findings, levels of 9-hydroxyoctadecadienoic acid (9-HODE), a known PPARγ agonist, were significantly increased in infected NSCs. Likewise, exposure of uninfected NSCs to 9-HODE recapitulated the effect of infection on PPARγ activity. It also increased the rate of cells expressing the IE antigen in HCMV-infected NSCs. Further, we demonstrated that (1) pharmacological activation of ectopically expressed PPARγ was sufficient to induce impaired neuronogenesis of uninfected NSCs, (2) treatment of uninfected NSCs with 9-HODE impaired NSC differentiation and (3) treatment of HCMV-infected NSCs with the PPARγ inhibitor T0070907 restored a normal rate of differentiation. The role of PPARγ in the disease phenotype was strongly supported by the immunodetection of nuclear PPARγ in brain germinative zones of congenitally infected fetuses (N = 20), but not in control samples. Altogether, our findings reveal a key role for PPARγ in neurogenesis and in the pathophysiology of HCMV congenital infection. They also pave the way to the identification of PPARγ gene targets in the infected brain. Congenital infection by human cytomegalovirus (HCMV) might result in permanent neurological sequelae, including sensorineural deafness, cerebral palsies or devastating neurodevelopmental abnormalities. Infants with such sequelae represent about 0.1% of all live births (>5500 per year in the USA). Given the considerable health and societal burden, a better insight on disease pathogenesis is urgently needed to design new therapeutic or prognostic tools. Here, we studied the impact of HCMV on neuronal development, using human neural progenitors (NSC) as a disease model. In particular, we investigated the outcome of infection on Peroxisome Proliferator-Activated Receptor gamma (PPARγ, a key protein in the regulation of metabolism, inflammation and cell differentiation. We disclosed that HCMV infection strongly increases levels and activity of PPARγ in NSCs. In vitro experiments showed that PPARγ activity inhibits the differentiation of NSCs into neurons. We also found increased PPARγ expression in brains of in utero infected fetuses, but not in controls, suggesting that PPARγ is a key effector of HCMV infection also in vivo. Our study provides new insights on the pathogenesis of HCMV infection and paves the way to the discovery of PPARγ-related molecules secreted in the infected brain.
Collapse
Affiliation(s)
- Maude Rolland
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Xiaojun Li
- Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Yann Sellier
- Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Hélène Martin
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Teresa Perez-Berezo
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Benjamin Rauwel
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | | | - Bettina Bessières
- Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Jacqueline Aziza
- Département d'Anatomie Pathologique, IUCT-Oncopole, Toulouse, France
| | - Nicolas Cenac
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Minhua Luo
- Laboratory of Virology, Wuhan Institute of Virology, Chinese Academy of Sciences, Wuhan, China
| | - Charlotte Casper
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
- Neonatal Unit, Children’s Hospital, Toulouse, France
| | - Marc Peschanski
- I-STEM, INSERM U861, AFM, Evry, France
- CECS, UEVE U861, Evry, France
| | - Daniel Gonzalez-Dunia
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Marianne Leruez-Ville
- Hôpital Necker-Enfants Malades, Assistance Publique-Hôpitaux de Paris, Paris, France
- Université Paris Descartes, Sorbonne Paris Cité, Paris, France
| | - Christian Davrinche
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
| | - Stéphane Chavanas
- Centre de Physiopathologie Toulouse Purpan, INSERM UMR 1043, CNRS UMR 5282, Université Paul Sabatier, Toulouse, France
- * E-mail:
| |
Collapse
|
44
|
Bolijn S, Lucassen PJ. How the Body Talks to the Brain; Peripheral Mediators of Physical Activity-Induced Proliferation in the Adult Hippocampus. Brain Plast 2015; 1:5-27. [PMID: 29765833 PMCID: PMC5939189 DOI: 10.3233/bpl-150020] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
In the hippocampal dentate gyrus, stem cells maintain the capacity to produce new neurons into adulthood. These adult-generated neurons become fully functional and are incorporated into the existing hippocampal circuit. The process of adult neurogenesis contributes to hippocampal functioning and is influenced by various environmental, hormonal and disease-related factors. One of the most potent stimuli of neurogenesis is physical activity (PA). While the bodily and peripheral changes of PA are well known, e.g. in relation to diet or cardiovascular conditions, little is known about which of these also exert central effects on the brain. Here, we discuss PA-induced changes in peripheral mediators that can modify hippocampal proliferation, and address changes with age, sex or PA duration/intensity. Of the many peripheral factors known to be triggered by PA, serotonin, FGF-2, IGF-1, VEGF, β-endorphin and adiponectin are best known for their stimulatory effects on hippocampal proliferation. Interestingly, while age negatively affects hippocampal proliferation per se, also the PA-induced response to most of these peripheral mediators is reduced and particularly the response to IGF-1 and NPY strongly declines with age. Sex differences per se have generally little effects on PA-induced neurogenesis. Compared to short term exercise, long term PA may negatively affect proliferation, due to a parallel decline in FGF-2 and the β-endorphin receptor, and an activation of the stress system particularly during conditions of prolonged exercise but this depends on other variables as well and remains a matter of discussion. Taken together, of many possible mediators, serotonin, FGF-2, IGF-1, VEGF, β-endorphin and adiponectin are the ones that most strongly contribute to the central effects of PA on the hippocampus. For a subgroup of these factors, brain sensitivity and responsivity is reduced with age.
Collapse
Affiliation(s)
- Simone Bolijn
- Centre for Neuroscience, Swammerdam Institute of Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| | - Paul J Lucassen
- Centre for Neuroscience, Swammerdam Institute of Life Sciences, University of Amsterdam, Amsterdam, The Netherlands
| |
Collapse
|
45
|
Li G, Zhou L, Zhu Y, Wang C, Sha S, Xian X, Ji Y, Liu G, Chen L. Seipin knockout in mice impairs stem cell proliferation and progenitor cell differentiation in the adult hippocampal dentate gyrus via reduced levels of PPARγ. Dis Model Mech 2015; 8:1615-24. [PMID: 26398946 PMCID: PMC4728316 DOI: 10.1242/dmm.021550] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2015] [Accepted: 09/14/2015] [Indexed: 12/16/2022] Open
Abstract
The seipin gene (BSCL2) was originally identified in humans as a loss-of-function gene associated with congenital generalized lipodystrophy type 2 (CGL2). Neuronal seipin-knockout (seipin-nKO) mice display a depression-like phenotype with a reduced level of hippocampal peroxisome proliferator-activated receptor gamma (PPARγ). The present study investigated the influence of seipin deficiency on adult neurogenesis in the hippocampal dentate gyrus (DG) and the underlying mechanisms of the effects. We show that the proliferative capability of stem cells in seipin-nKO mice was substantially reduced compared to in wild-type (WT) mice, and that this could be rescued by the PPARγ agonist rosiglitazone (rosi). In seipin-nKO mice, neuronal differentiation of progenitor cells was inhibited, with the enhancement of astrogliogenesis; both of these effects were recovered by rosi treatment during early stages of progenitor cell differentiation. In addition, rosi treatment could correct the decline in hippocampal ERK2 phosphorylation and cyclin A mRNA level in seipin-nKO mice. The MEK inhibitor U0126 abolished the rosi-rescued cell proliferation and cyclin A expression in seipin-nKO mice. In seipin-nKO mice, the hippocampal Wnt3 protein level was less than that in WT mice, and there was a reduction of neurogenin 1 (Neurog1) and neurogenic differentiation 1 (NeuroD1) mRNA, levels of which were corrected by rosi treatment. STAT3 phosphorylation (Tyr705) was enhanced in seipin-nKO mice, and was further elevated by rosi treatment. Finally, rosi treatment for 10 days could alleviate the depression-like phenotype in seipin-nKO mice, and this alleviation was blocked by the MEK inhibitor U0126. The results indicate that, by reducing PPARγ, seipin deficiency impairs proliferation and differentiation of neural stem and progenitor cells, respectively, in the adult DG, which might be responsible for the production of the depression-like phenotype in seipin-nKO mice.
Collapse
Affiliation(s)
- Guoxi Li
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 210029, China Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | - Libin Zhou
- Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | - Ying Zhu
- Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | - Conghui Wang
- Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | - Sha Sha
- Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| | - Xunde Xian
- Department of Molecular Genetics, UT Southwestern Medical Center, Dallas, TX 75390, USA
| | - Yong Ji
- Key Laboratory of Cardiovascular Disease and Molecular Intervention, Atherosclerosis Research Centre, Nanjing Medical University, Nanjing 210029, China
| | - George Liu
- Institute of Cardiovascular Sciences, Peking University, Beijing 100191, China
| | - Ling Chen
- State Key Laboratory of Reproductive Medicine, Nanjing Medical University, Nanjing 210029, China Department of Physiology, Nanjing Medical University, Nanjing 210029, China
| |
Collapse
|
46
|
Liu Y, Huang Y, Lee S, Bookout AL, Castorena CM, Wu H, Gautron L. PPARγ mRNA in the adult mouse hypothalamus: distribution and regulation in response to dietary challenges. Front Neuroanat 2015; 9:120. [PMID: 26388745 PMCID: PMC4558427 DOI: 10.3389/fnana.2015.00120] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/03/2015] [Accepted: 08/20/2015] [Indexed: 11/13/2022] Open
Abstract
Peroxisome proliferator-activated receptor gamma (PPARγ) is a ligand-activated transcription factor that was originally identified as a regulator of peroxisome proliferation and adipocyte differentiation. Emerging evidence suggests that functional PPARγ signaling also occurs within the hypothalamus. However, the exact distribution and identities of PPARγ-expressing hypothalamic cells remains under debate. The present study systematically mapped PPARγ mRNA expression in the adult mouse brain using in situ hybridization histochemistry. PPARγ mRNA was found to be expressed at high levels outside the hypothalamus including the neocortex, the olfactory bulb, the organ of the vasculosum of the lamina terminalis (VOLT), and the subfornical organ. Within the hypothalamus, PPARγ was present at moderate levels in the suprachiasmatic nucleus (SCh) and the ependymal of the 3rd ventricle. In all examined feeding-related hypothalamic nuclei, PPARγ was expressed at very low levels that were close to the limit of detection. Using qPCR techniques, we demonstrated that PPARγ mRNA expression was upregulated in the SCh in response to fasting. Double in situ hybridization further demonstrated that PPARγ was primarily expressed in neurons rather than glia. Collectively, our observations provide a comprehensive map of PPARγ distribution in the intact adult mouse hypothalamus.
Collapse
Affiliation(s)
- Yang Liu
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center Dallas, TX, USA ; Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Ying Huang
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center Dallas, TX, USA
| | - Syann Lee
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center Dallas, TX, USA
| | - Angie L Bookout
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center Dallas, TX, USA
| | - Carlos M Castorena
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center Dallas, TX, USA
| | - Hua Wu
- Department of Orthopedics, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology Wuhan, China
| | - Laurent Gautron
- Division of Hypothalamic Research, Department of Internal Medicine, University of Texas Southwestern Medical Center Dallas, TX, USA
| |
Collapse
|
47
|
Taheri M, Salamian A, Ghaedi K, Peymani M, Izadi T, Nejati AS, Atefi A, Nematollahi M, Ahmadi Ghahrizjani F, Esmaeili M, Kiani Esfahani A, Irani S, Baharvand H, Nasr-Esfahani MH. A ground state of PPARγ activity and expression is required for appropriate neural differentiation of hESCs. Pharmacol Rep 2015; 67:1103-14. [PMID: 26481528 DOI: 10.1016/j.pharep.2015.04.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2014] [Revised: 04/14/2015] [Accepted: 04/17/2015] [Indexed: 01/21/2023]
Abstract
BACKGROUND Several evidences indicate stimulation of peroxisome proliferator activated receptor γ (PPARg), promotes neuronal differentiation. This study was conducted to testify the prominence of PPARγ during neural differentiation of human embryonic stem cells (hESCs). METHODS PPARγ expression level was assessed during neural differentiation of hESCs. Meanwhile, the level of endogenous miRNAs, which could be engaged in regulation of PPARγ expression, was measured. Next, natural and synthetic components of PPARγ agonists and antagonist were implemented on neural progenitor formation during neural differentiation of hESCs. RESULTS Data showed an increasing wave of PPARγ expression level when human neural progenitors (NPs) were formed upon retinoic acid treatment. Interestingly, there was no significant difference in the amount of PPARγ proteins during the differentiation of hESCs that is inconsistent with what we observed for RNA level. Our results indicated that miRNAs are not involved in the regulation of PPARγ expression, while proteasome-mediated degradation may to some degree be involved in this process. Among numerous treatments, PPARγ inactivation during NPs formation significantly decreased expression of NP markers. CONCLUSIONS We conclude that a ground state of PPARγ activity is required for NP formation of hESCs during early neural differentiation. However, high expression and activity of PPARγ could not enhance the required neural differentiation, whereas the PPARγ inactivation could negatively influence NP formation from hESCs by antagonist.
Collapse
Affiliation(s)
- Marjan Taheri
- Biology Department, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Ahmad Salamian
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Kamran Ghaedi
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran; Department of Biology, School of Sciences, University of Isfahan, Isfahan, Iran.
| | - Maryam Peymani
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Tayebeh Izadi
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Alireza Shoaraye Nejati
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Atefeh Atefi
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Marzieh Nematollahi
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Fatemeh Ahmadi Ghahrizjani
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Maryam Esmaeili
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Abbas Kiani Esfahani
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran
| | - Shiva Irani
- Biology Department, Science and Research Branch, Islamic Azad University, Tehran, Iran
| | - Hossein Baharvand
- Department of Stem Cells and Developmental Biology at Cell Science Research Center, Royan Institute for Stem Cell Biology and Technology, ACECR, Tehran, Iran; Department of Developmental Biology, University of Science and Culture, ACECR, Tehran, Iran
| | - Mohammad Hossein Nasr-Esfahani
- Department of Cellular Biotechnology at Cell Science Research Center, Royan Institute for Biotechnology, ACECR, Isfahan, Iran.
| |
Collapse
|
48
|
Bernal C, Araya C, Palma V, Bronfman M. PPARβ/δ and PPARγ maintain undifferentiated phenotypes of mouse adult neural precursor cells from the subventricular zone. Front Cell Neurosci 2015; 9:78. [PMID: 25852474 PMCID: PMC4364249 DOI: 10.3389/fncel.2015.00078] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2014] [Accepted: 02/21/2015] [Indexed: 12/13/2022] Open
Abstract
The subventricular zone (SVZ) is one of the main niches of neural stem cells in the adult mammalian brain. Stem and precursor cells in this region are the source for neurogenesis and oligodendrogesis, mainly in the olfactory bulb and corpus callosum, respectively. The identification of the molecular components regulating the decision of these cells to differentiate or maintain an undifferentiated state is important in order to understand the modulation of neurogenic processes in physiological and pathological conditions. PPARs are a group of transcription factors, activated by lipid ligands, with important functions in cellular differentiation and proliferation in several tissues. In this work, we demonstrate that mouse adult neural precursor cells (NPCs), in situ and in vitro, express PPARβ/δ and PPARγ. Pharmacological activation of both PPARs isoforms induces proliferation and maintenance of the undifferentiated phenotype. Congruently, inhibition of PPARβ/δ and PPARγ results in a decrease of proliferation and loss of the undifferentiated phenotype. Interestingly, PPARγ regulates the level of EGFR in adult NPCs, concurrent with it is function described in embryonic NPCs. Furthermore, we describe for the first time that PPARβ/δ regulates SOX2 level in adult NPCs, probably through a direct transcriptional regulation, as we identified two putative PPAR response elements in the promoter region of Sox2. EGFR and SOX2 are key players in neural stem/precursor cells self-renewal. Finally, rosiglitazone, a PPARγ ligand, increases PPARβ/δ level, suggesting a possible cooperation between these two PPARs in the control of cell fate behavior. Our work contributes to the understanding of the molecular mechanisms associated to neural cell fate decision and places PPARβ/δ and PPARγ as interesting new targets of modulation of mammalian brain homeostasis.
Collapse
Affiliation(s)
- Carolina Bernal
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Center for Aging and Regeneration, Pontifical Catholic University of Chile Santiago, Chile
| | - Claudia Araya
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Center for Aging and Regeneration, Pontifical Catholic University of Chile Santiago, Chile
| | - Verónica Palma
- Laboratory of Stem Cells and Development, Faculty of Science, FONDAP Center for Genome Regulation, University of Chile Santiago, Chile
| | - Miguel Bronfman
- Department of Cell and Molecular Biology, Faculty of Biological Sciences, Center for Aging and Regeneration, Pontifical Catholic University of Chile Santiago, Chile
| |
Collapse
|
49
|
Zhu JM, Hu N. Expression of peroxisome proliferator-activated receptor γ in rat retina during development. Int J Ophthalmol 2015; 8:52-6. [PMID: 25709907 DOI: 10.3980/j.issn.2222-3959.2015.01.09] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2014] [Accepted: 10/29/2014] [Indexed: 11/02/2022] Open
Abstract
AIM To evaluate the spatiotemporal expression pattern of PPARγ in embryonic and early postnatal stages of rat retina. METHODS Fetal rats were collected at 13-18d of gestation (GD) from pregnant females and postnatal rats at 1d (P1) and 5d (P5) after birth were also used. We used RT-PCR to detect PPARγ mRNA and immunohistochemical to observe PPARγ protein. And at last, we chose HE staining showed the structural changes of rat retina during development. RESULTS RT-PCR analysis showed that PPARγ mRNA was expressed as early as GD13 and gradually decreased as maturation continued. However, the PPARγ gene expression significantly increased after birth, especially in P5. Immunohistochemical analysis showed PPARγ protein was expressed throughout the retinal neuroepithelium at GD13 and GD14, and then decreased during late embryogenesis but remained relatively high in the predicted ganglion cell zone. During postnatal development, PPARγ protein was remarkably increased and the positive signals were mainly located in nerve fiber layer (NFL), ganglion cell layer (GCL) and outer layers of the retina. CONCLUSION The spatiotemporal changes of PPARγ expression demonstrated that PPARγ might play a role in regulating the differentiation and maturation of retinal cells.
Collapse
Affiliation(s)
- Ju-Ming Zhu
- The Fourth Affiliated Hospital of Nantong Medical College, Yancheng City No.1 People's Hospital, Yancheng 224006, Jiangsu Province, China
| | - Nan Hu
- Eye Institute, Affiliated Hospital of Nantong University, Nantong 226001, Jiangsu Province, China
| |
Collapse
|
50
|
Crenshaw DG, Asin K, Gottschalk WK, Liang Z, Zhang N, Roses AD. Effects of low doses of pioglitazone on resting-state functional connectivity in conscious rat brain. PLoS One 2015; 10:e0117973. [PMID: 25671601 PMCID: PMC4324644 DOI: 10.1371/journal.pone.0117973] [Citation(s) in RCA: 16] [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: 05/06/2014] [Accepted: 12/26/2014] [Indexed: 12/25/2022] Open
Abstract
Pioglitazone (PIO) is a peroxisome proliferator-activated receptor-γ (PPARγ) agonist in clinical use for treatment of type 2 diabetes (T2DM). Accumulating evidence suggests PPARγ agonists may be useful for treating or delaying the onset of Alzheimer's disease (AD), possibly via actions on mitochondria, and that dose strengths lower than those clinically used for T2DM may be efficacious. Our major objective was to determine if low doses of pioglitazone, administered orally, impacted brain activity. We measured blood-oxygenation-level dependent (BOLD) low-frequency fluctuations in conscious rats to map changes in brain resting-state functional connectivity due to daily, oral dosing with low-dose PIO. The connectivity in two neural circuits exhibited significant changes compared with vehicle after two days of treatment with PIO at 0.08 mg/kg/day. After 7 days of treatment with a range of PIO dose-strengths, connections between 17 pairs of brain regions were significantly affected. Functional connectivity with the CA1 region of the hippocampus, a region that is involved in memory and is affected early in the progression of AD, was specifically investigated in a seed-based analysis. This approach revealed that the spatial pattern of CA1 connectivity was consistent among all dose groups at baseline, prior to treatment with PIO, and in the control group imaged on day 7. Compared to baseline and controls, increased connectivity to CA1 was observed regionally in the hypothalamus and ventral thalamus in all PIO-treated groups, but was least pronounced in the group treated with the highest dose of PIO. These data support our hypothesis that PIO modulates neuronal and/or cerebrovascular function at dose strengths significantly lower than those used to treat T2DM and therefore may be a useful therapy for neurodegenerative diseases including AD.
Collapse
Affiliation(s)
- Donna G. Crenshaw
- Zinfandel Pharmaceuticals, Inc., Chapel Hill, NC, United States of America
| | - Karen Asin
- Takeda Development Center Americas, Inc., Deerfield, IL, United States of America
| | | | - Zhifeng Liang
- Biomedical Engineering Department, The Pennsylvania State University, University Park, PA, United States of America
| | - Nanyin Zhang
- Biomedical Engineering Department, The Pennsylvania State University, University Park, PA, United States of America
| | - Allen D. Roses
- Zinfandel Pharmaceuticals, Inc., Chapel Hill, NC, United States of America
| |
Collapse
|