1
|
Zhu C, Wang L, Nie X, Yang X, Gao K, Jiang Z. Dietary dibutyryl cAMP supplementation regulates the fat deposition in adipose tissues of finishing pigs via cAMP/PKA pathway. Anim Biotechnol 2023; 34:921-934. [PMID: 34871537 DOI: 10.1080/10495398.2021.2003373] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
This study investigated potential mechanism of dibutyryl-cAMP (db-cAMP) on porcine fat deposition. (1) Exp.1, 72 finishing pigs were allotted to 3 treatments (0, 10 or 20 mg/kg dbcAMP) with 6 replicates. dbcAMP increased the hormone sensitive lipase (HSL) activity and expression of β-adrenergic receptor (β-AR) and growth hormone receptor (GHR), but decreased expression of peroxisome proliferator-activated receptor gamma 2 (PPAR-γ2) and adipocyte fatty acid binding protein (A-FABP) in back fat. dbcAMP upregulated expression of β-AR, GHR, PPAR-γ2 and A-FABP, but decreased insulin receptor (INSR) expression in abdominal fat. Dietary dbcAMP increased HSL activity and expression of G protein-coupled receptor (GPCR), cAMP-response element-binding protein (CREB) and insulin-like growth factor-1 (IGF-1), but decreased fatty acid synthase (FAS) and lipoprotein lipase (LPL) activities, and expression of INSR, cAMP-response element-binding protein (C/EBP-α) and A-FABP in perirenal fat. (2) Exp. 2, dbcAMP suppressed the proliferation and differentiation of porcine preadipocytes in a time- and dose-dependent manner, which might be associated with increased activities of cAMP and protein kinase A (PKA), and expression of GPCR, β-AR, GHR and CREB via inhibiting C/EBP-α and PPAR-γ2 expression. Collectively, dbcAMP treatment may reduce fat deposition by regulating gene expression related to adipocyte differentiation and fat metabolism partially via cAMP-PKA pathway.
Collapse
Affiliation(s)
- Cui Zhu
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Li Wang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Xiaoyan Nie
- School of Life Science and Engineering, Foshan University, Foshan, China
| | - Xuefen Yang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Kaiguo Gao
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| | - Zongyong Jiang
- Institute of Animal Science, Guangdong Academy of Agricultural Sciences, State Key Laboratory of Livestock and Poultry Breeding, Ministry of Agriculture Key Laboratory of Animal Nutrition and Feed Science in South China, Guangdong Provincial Key Laboratory of Animal Breeding and Nutrition, Maoming Branch, Guangdong Laboratory for Lingnan Modern Agriculture, Guangzhou, China
| |
Collapse
|
2
|
Belo do Nascimento I, Verfaillie M, Ates G, Beckers P, Joris V, Desmet N, Massie A, Hermans E. AMPK Modulates the Metabolic Adaptation of C6 Glioma Cells in Glucose-Deprived Conditions without Affecting Glutamate Transport. Cells 2022; 11:cells11111800. [PMID: 35681495 PMCID: PMC9180554 DOI: 10.3390/cells11111800] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2022] [Revised: 05/26/2022] [Accepted: 05/26/2022] [Indexed: 02/04/2023] Open
Abstract
Energy homeostasis in the central nervous system largely depends on astrocytes, which provide metabolic support and protection to neurons. Astrocytes also ensure the clearance of extracellular glutamate through high-affinity transporters, which indirectly consume ATP. Considering the role of the AMP-activated protein kinase (AMPK) in the control of cell metabolism, we have examined its implication in the adaptation of astrocyte functions in response to a metabolic stress triggered by glucose deprivation. We genetically modified the astrocyte-like C6 cell line to silence AMPK activity by overexpressing a dominant negative mutant of its catalytic subunit. Upon glucose deprivation, we found that C6 cells maintain stable ATP levels and glutamate uptake capacity, highlighting their resilience during metabolic stress. In the same conditions, cells with silenced AMPK activity showed a reduction in motility, metabolic activity, and ATP levels, indicating that their adaptation to stress is compromised. The rate of ATP production remained, however, unchanged by AMPK silencing, suggesting that AMPK mostly influences energy consumption during stress conditions in these cells. Neither AMPK modulation nor prolonged glucose deprivation impaired glutamate uptake. Together, these results indicate that AMPK contributes to the adaptation of astrocyte metabolism triggered by metabolic stress, but not to the regulation of glutamate transport.
Collapse
Affiliation(s)
- Inês Belo do Nascimento
- Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium; (I.B.d.N.); (M.V.); (P.B.); (N.D.)
| | - Marie Verfaillie
- Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium; (I.B.d.N.); (M.V.); (P.B.); (N.D.)
| | - Gamze Ates
- Center for Neurosciences, Neuro-Aging & Viro-Immunotherapy, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (G.A.); (A.M.)
| | - Pauline Beckers
- Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium; (I.B.d.N.); (M.V.); (P.B.); (N.D.)
| | - Virginie Joris
- Pole of Pharmacology and Therapeutics, Institute of Experimental and Clinical Research (IREC), Université Catholique de Louvain (UCLouvain), 1200 Brussels, Belgium;
| | - Nathalie Desmet
- Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium; (I.B.d.N.); (M.V.); (P.B.); (N.D.)
| | - Ann Massie
- Center for Neurosciences, Neuro-Aging & Viro-Immunotherapy, Vrije Universiteit Brussel (VUB), 1090 Brussels, Belgium; (G.A.); (A.M.)
| | - Emmanuel Hermans
- Institute of Neuroscience, Université Catholique de Louvain, 1200 Brussels, Belgium; (I.B.d.N.); (M.V.); (P.B.); (N.D.)
- Correspondence: ; Tel.: +32-2764-9339
| |
Collapse
|
3
|
Chen Z, Zhong Y, Chen J, Sun S, Liu W, Han Y, Liu X, Guo C, Li D, Hu W, Zhang P, Chen Z, Chen Z, Mou Y, Yan G, Zhu W, Yin W, Sai K. Disruption of β-catenin-mediated negative feedback reinforces cAMP-induced neuronal differentiation in glioma stem cells. Cell Death Dis 2022; 13:493. [PMID: 35610201 PMCID: PMC9130142 DOI: 10.1038/s41419-022-04957-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2021] [Revised: 05/11/2022] [Accepted: 05/16/2022] [Indexed: 12/14/2022]
Abstract
Accumulating evidence supports the existence of glioma stem cells (GSCs) and their critical role in the resistance to conventional treatments for glioblastoma multiforme (GBM). Differentiation therapy represents a promising alternative strategy against GBM by forcing GSCs to exit the cell cycle and reach terminal differentiation. In this study, we demonstrated that cAMP triggered neuronal differentiation and compromised the self-renewal capacity in GSCs. In addition, cAMP induced negative feedback to antagonize the differentiation process by activating β-catenin pathway. Suppression of β-catenin signaling synergized with cAMP activators to eliminate GSCs in vitro and extended the survival of animals in vivo. The cAMP/PKA pathway stabilized β-catenin through direct phosphorylation of the molecule and inhibition of GSK-3β. The activated β-catenin translocated into the nucleus and promoted the transcription of APELA and CARD16, which were found to be responsible for the repression of cAMP-induced differentiation in GSCs. Overall, our findings identified a negative feedback mechanism for cAMP-induced differentiation in GSCs and provided potential targets for the reinforcement of differentiation therapy for GBM.
Collapse
Affiliation(s)
- Zhijie Chen
- grid.488530.20000 0004 1803 6191Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China ,grid.412558.f0000 0004 1762 1794Department of Neurosurgery, The Third Affiliated Hospital of Sun Yat-sen University Lingnan Hospital, Guangzhou, 510530 China
| | - Yingqian Zhong
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 China
| | - Jiehong Chen
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 China
| | - Shuxin Sun
- grid.410643.4Department of Pancreas Center, Guangdong Provincial People’s Hospital, Guangdong Academy of Medical Sciences, Guangzhou, 510080 China
| | - Wenfeng Liu
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 China
| | - Yu Han
- grid.488530.20000 0004 1803 6191Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China
| | - Xincheng Liu
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 China
| | - Cui Guo
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 China
| | - Depei Li
- grid.488530.20000 0004 1803 6191Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China
| | - Wanming Hu
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China ,grid.488530.20000 0004 1803 6191Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China
| | - Peiyu Zhang
- grid.488530.20000 0004 1803 6191Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China
| | - Zhuopeng Chen
- grid.488530.20000 0004 1803 6191Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China
| | - Zhongping Chen
- grid.488530.20000 0004 1803 6191Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China
| | - Yonggao Mou
- grid.488530.20000 0004 1803 6191Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China
| | - Guangmei Yan
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 China
| | - Wenbo Zhu
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 China
| | - Wei Yin
- grid.12981.330000 0001 2360 039XDepartment of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, 510080 China
| | - Ke Sai
- grid.488530.20000 0004 1803 6191Department of Neurosurgery/Neuro-oncology, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China ,grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Sun Yat-sen University Cancer Center, Guangzhou, 510060 China
| |
Collapse
|
4
|
Chonan Y, Yamashita T, Sampetrean O, Saya H, Sudo R. Spatial heterogeneity of invading glioblastoma cells regulated by paracrine factors. Tissue Eng Part A 2021; 28:573-585. [PMID: 34841881 DOI: 10.1089/ten.tea.2021.0168] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common and lethal type of malignant primary brain tumor in adults. GBM displays heterogeneous tumor cell population comprising glioma-initiating cells (GICs) with stem cell-like characteristics and differentiated glioma cells. During GBM cell invasion into normal brain tissues, which is the hallmark characteristic of GBM, GICs at the invasion front retain stemness, while cells at the tumor core display cellular differentiation. However, the mechanism of cellular differentiation underlying the formation of spatial cellular heterogeneity in GBM remains unknown. In the present study, we first observed spatially heterogeneous GBM cell populations emerged from an isogenic clonal population of GICs during invasion into a 3D collagen hydrogel in a microfluidic device. Specifically, GICs at the invasion front maintained stemness, while trailing cells displayed astrocytic differentiation. The spatial cellular heterogeneity resulted from the difference in cell density between GICs at the invasion front and trailing cells. Trailing GICs at high cell density exhibited astrocytic differentiation via local accumulation of paracrine factors they secreted, while cells at the invasion front of low cell density retained stemness due to the lack of paracrine factors. In addition, we demonstrated that interstitial flow suppressed astrocytic differentiation of trailing GICs by the clearance of paracrine factors. Our findings suggest that intercellular crosstalk between tumor cells is an essential factor in developing the spatial cellular heterogeneity of GBM cells with various differentiation statuses. It also provides insights into the development of novel therapeutic strategies targeting GBM cells with stem cell characteristics at the invasion front.
Collapse
Affiliation(s)
- Yuta Chonan
- Keio University, School of Integrated Design Engineering, Yokohama, Kanagawa, Japan;
| | - Tadahiro Yamashita
- Keio University, Department of System Design Engineering, Yokohama, Kanagawa, Japan.,Keio University, School of Integrated Design Engineering, Yokohama, Kanagawa, Japan;
| | - Oltea Sampetrean
- Keio University School of Medicine, Division of Gene Regulation, Institute for Advanced Medical Research, Tokyo, Japan;
| | - Hideyuki Saya
- Keio University School of Medicine, Division of Gene Regulation, Institute for Advanced Medical Research, Tokyo, Japan;
| | - Ryo Sudo
- Keio University, Department of System Design Engineering, Yokohama, Kanagawa, Japan.,Keio University, School of Integrated Design Engineering, Yokohama, Kanagawa, Japan;
| |
Collapse
|
5
|
Guijarro-Belmar A, Domanski DM, Bo X, Shewan D, Huang W. The therapeutic potential of targeting exchange protein directly activated by cyclic adenosine 3',5'-monophosphate (Epac) for central nervous system trauma. Neural Regen Res 2021; 16:460-469. [PMID: 32985466 PMCID: PMC7996029 DOI: 10.4103/1673-5374.293256] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
Millions of people worldwide are affected by traumatic spinal cord injury, which usually results in permanent sensorimotor disability. Damage to the spinal cord leads to a series of detrimental events including ischaemia, haemorrhage and neuroinflammation, which over time result in further neural tissue loss. Eventually, at chronic stages of traumatic spinal cord injury, the formation of a glial scar, cystic cavitation and the presence of numerous inhibitory molecules act as physical and chemical barriers to axonal regrowth. This is further hindered by a lack of intrinsic regrowth ability of adult neurons in the central nervous system. The intracellular signalling molecule, cyclic adenosine 3′,5′-monophosphate (cAMP), is known to play many important roles in the central nervous system, and elevating its levels as shown to improve axonal regeneration outcomes following traumatic spinal cord injury in animal models. However, therapies directly targeting cAMP have not found their way into the clinic, as cAMP is ubiquitously present in all cell types and its manipulation may have additional deleterious effects. A downstream effector of cAMP, exchange protein directly activated by cAMP 2 (Epac2), is mainly expressed in the adult central nervous system, and its activation has been shown to mediate the positive effects of cAMP on axonal guidance and regeneration. Recently, using ex vivo modelling of traumatic spinal cord injury, Epac2 activation was found to profoundly modulate the post-lesion environment, such as decreasing the activation of astrocytes and microglia. Pilot data with Epac2 activation also suggested functional improvement assessed by in vivo models of traumatic spinal cord injury. Therefore, targeting Epac2 in traumatic spinal cord injury could represent a novel strategy in traumatic spinal cord injury repair, and future work is needed to fully establish its therapeutic potential.
Collapse
Affiliation(s)
- Alba Guijarro-Belmar
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen; Sainsbury Wellcome Centre, University College London, London, UK
| | - Dominik Mateusz Domanski
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen, UK
| | - Xuenong Bo
- Center for Neuroscience, Surgery and Trauma, Queen Mary University of London, London, UK
| | - Derryck Shewan
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen, UK
| | - Wenlong Huang
- Institute of Medical Sciences, School of Medicine, Medical Sciences & Nutrition, University of Aberdeen, Aberdeen, UK
| |
Collapse
|
6
|
Differentiation of Tumorigenic C6 Glioma Cells Induced by Enhanced IL-6 Signaling. ACTA ACUST UNITED AC 2020; 56:medicina56110625. [PMID: 33227992 PMCID: PMC7699282 DOI: 10.3390/medicina56110625] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2020] [Revised: 11/13/2020] [Accepted: 11/17/2020] [Indexed: 12/24/2022]
Abstract
Background and objectives: Cancer stem cells (CSCs) are obstacles to cancer therapy due to their therapeutic resistance, ability to initiate neoplasia, and roles in tumor relapse and metastasis. Efforts have been made to cure CSCs, such as the use of differentiation therapy, which induces cancer stem-like cells to undergo differentiation and decrease their tumorigenicity. Interleukin 6 (IL-6) upregulates the expression of glial fibrillary acidic protein (GFAP) in C6 glioma cells, indicating that it is able to induce the differentiation of these cells. The C6 glioma cell line forms a high percentage of cancer stem-like cells, leading us to speculate whether IL-6 signaling could modulate the differentiation of tumorigenic C6 glioma cells. However, we observed that IL-6 alone could not efficiently induce the differentiation of these cells. Therefore, different IL-6 signaling elicitors, including IL-6 alone, a combination of IL-6 and soluble IL-6 receptor (IL-6/sIL-6R), and tumor necrosis factor-α (TNF-α) plus IL-6/sIL-6R (TNF-α/IL-6/sIL-6R), were evaluated for their potential use in differentiation therapy. Materials and Methods: The potential of IL-6 signaling elicitors in differentiation therapy were examined by assessing changes in biomarker levels, the rate of cell proliferation, and tumorigenicity, respectively. Results: Enhanced IL-6 signaling could effectively induce C6 glioma cell differentiation, as determined by observed variations in the expression of differentiation, cell cycle, and stem cell biomarkers. Additionally, the total cell population and the tumorigenicity of glioma cells were all considerably reduced after TNF-α/IL-6/sIL-6R treatment. Conclusions: Our findings provide evidence that enhanced IL-6 signaling can efficiently promote tumorigenic C6 glioma cells to undergo differentiation.
Collapse
|
7
|
Muniroh M. Methylmercury-induced pro-inflammatory cytokines activation and its preventive strategy using anti-inflammation N-acetyl-l-cysteine: a mini-review. REVIEWS ON ENVIRONMENTAL HEALTH 2020; 35:233-238. [PMID: 32710722 DOI: 10.1515/reveh-2020-0026] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/24/2020] [Accepted: 06/09/2020] [Indexed: 06/11/2023]
Abstract
The exposure of methylmercury (MeHg) has become a public health concern because of its neurotoxic effect. Various neurological symptoms were detected in Minamata disease patients, who got intoxicated by MeHg, including paresthesia, ataxia, gait disturbance, sensory disturbances, tremors, visual, and hearing impairments, indicating that MeHg could pass the blood-brain barrier (BBB) and cause impairment of neurons and other brain cells. Previous studies have reported some expected mechanisms of MeHg-induced neurotoxicity including the neuroinflammation pathway. It was characterized by the up-regulation of numerous pro-inflammatory cytokines expression. Therefore, the use of anti-inflammatories such as N-acetyl-l-cysteine (NAC) may act as a preventive compound to protect the brain from MeHg harmful effects. This mini-review will explain detailed information on MeHg-induced pro-inflammatory cytokines activation as well as possible preventive strategies using anti-inflammation NAC to protect brain cells, particularly in in vivo and in vitro studies.
Collapse
Affiliation(s)
- Muflihatul Muniroh
- Department of Physiology, Faculty of Medicine Diponegoro University, Semarang, Central Java, 50275, Indonesia
| |
Collapse
|
8
|
Pérez-Pérez D, Reyes-Vidal I, Chávez-Cortez EG, Sotelo J, Magaña-Maldonado R. Methylxanthines: Potential Therapeutic Agents for Glioblastoma. Pharmaceuticals (Basel) 2019; 12:ph12030130. [PMID: 31500285 PMCID: PMC6789489 DOI: 10.3390/ph12030130] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2019] [Revised: 08/24/2019] [Accepted: 09/01/2019] [Indexed: 12/13/2022] Open
Abstract
Glioblastoma (GBM) is the most common and aggressive primary brain tumor. Currently, treatment is ineffective and the median overall survival is 20.9 months. The poor prognosis of GBM is a consequence of several altered signaling pathways that favor the proliferation and survival of neoplastic cells. One of these pathways is the deregulation of phosphodiesterases (PDEs). These enzymes participate in the development of GBM and may have value as therapeutic targets to treat GBM. Methylxanthines (MXTs) such as caffeine, theophylline, and theobromine are PDE inhibitors and constitute a promising therapeutic anti-cancer agent against GBM. MTXs also regulate various cell processes such as proliferation, migration, cell death, and differentiation; these processes are related to cancer progression, making MXTs potential therapeutic agents in GBM.
Collapse
Affiliation(s)
- Daniel Pérez-Pérez
- PECEM, Faculty of Medicine, National Autonomous University of México, México City 04510, Mexico
- Neuroimmunology and Neuro-oncology Unit, National Institute of Neurology and Neurosurgery, México City 14269, Mexico
| | - Iannel Reyes-Vidal
- Neuroimmunology and Neuro-oncology Unit, National Institute of Neurology and Neurosurgery, México City 14269, Mexico
| | - Elda Georgina Chávez-Cortez
- Neuroimmunology and Neuro-oncology Unit, National Institute of Neurology and Neurosurgery, México City 14269, Mexico
| | - Julio Sotelo
- Neuroimmunology and Neuro-oncology Unit, National Institute of Neurology and Neurosurgery, México City 14269, Mexico
| | - Roxana Magaña-Maldonado
- Neuroimmunology and Neuro-oncology Unit, National Institute of Neurology and Neurosurgery, México City 14269, Mexico.
| |
Collapse
|
9
|
Epac2 Elevation Reverses Inhibition by Chondroitin Sulfate Proteoglycans In Vitro and Transforms Postlesion Inhibitory Environment to Promote Axonal Outgrowth in an Ex Vivo Model of Spinal Cord Injury. J Neurosci 2019; 39:8330-8346. [PMID: 31409666 DOI: 10.1523/jneurosci.0374-19.2019] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 08/05/2019] [Accepted: 08/08/2019] [Indexed: 02/07/2023] Open
Abstract
Millions of patients suffer from debilitating spinal cord injury (SCI) without effective treatments. Elevating cAMP promotes CNS neuron growth in the presence of growth-inhibiting molecules. cAMP's effects on neuron growth are partly mediated by Epac, comprising Epac1 and Epac2; the latter predominantly expresses in postnatal neural tissue. Here, we hypothesized that Epac2 activation would enhance axonal outgrowth after SCI. Using in vitro assays, we demonstrated, for the first time, that Epac2 activation using a specific soluble agonist (S-220) significantly enhanced neurite outgrowth of postnatal rat cortical neurons and markedly overcame the inhibition by chondroitin sulfate proteoglycans and mature astrocytes on neuron growth. We further investigated the novel potential of Epac2 activation in promoting axonal outgrowth by an ex vivo rat model of SCI mimicking post-SCI environment in vivo and by delivering S-220 via a self-assembling Fmoc-based hydrogel that has suitable properties for SCI repair. We demonstrated that S-220 significantly enhanced axonal outgrowth across the lesion gaps in the organotypic spinal cord slices, compared with controls. Furthermore, we elucidated, for the first time, that Epac2 activation profoundly modulated the lesion environment by reducing astrocyte/microglial activation and transforming astrocytes into elongated morphology that guided outgrowing axons. Finally, we showed that S-220, when delivered by the gel at 3 weeks after contusion SCI in male adult rats, resulted in significantly better locomotor performance for up to 4 weeks after treatment. Our data demonstrate a promising therapeutic potential of S-220 in SCI, via beneficial effects on neurons and glia after injury to facilitate axonal outgrowth.SIGNIFICANCE STATEMENT During development, neuronal cAMP levels decrease significantly compared with the embryonic stage when the nervous system is established. This has important consequences following spinal cord injury, as neurons fail to regrow. Elevating cAMP levels encourages injured CNS neurons to sprout and extend neurites. We have demonstrated that activating its downstream effector, Epac2, enhances neurite outgrowth in vitro, even in the presence of an inhibitory environment. Using a novel biomaterial-based drug delivery system in the form of a hydrogel to achieve local delivery of an Epac2 agonist, we further demonstrated that specific activation of Epac2 enhances axonal outgrowth and minimizes glial activation in an ex vivo model of spinal cord injury, suggesting a new strategy for spinal cord repair.
Collapse
|
10
|
Hisab AS. Effects of cyclic AMP on the differentiation and bioenergetics of rat C6 glioma cells. Int J Neurosci 2018; 129:230-244. [PMID: 30232914 DOI: 10.1080/00207454.2018.1526798] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022]
Abstract
INTRODUCTION Elevation in the level of intracellular cAMP is known to induce astrocytic differentiation of C6 glioma cells by unknown mechanisms. METHODS Therefore, cytoskeletal protein genes (phalloidin) fluorescents to investigate morphological changes, cell proliferation assay, MTT assay, flow cytometry, western blotting, in-cell western, immune-cytochemical (protein expression and localization), and oxygen electrodes (oxygen consumption rate) after a treatment with 0.25 mM dbcAMP were conducted. RESULTS Undifferentiated cells (media without dbcAMP) showed a flat polygonal appearance, whereas those cultured in the presence of 0.25 mM dbcAMP exhibited a more differentiated astrocytic morphology. They had more numerous neurite-like thin processes. The cell proliferation of differentiated c6 glioma reduced at day 2 and then started to increase at day 3 till day 5 compared to undifferentiated c6 glioma cells. In terms of flow-cytometry data, dbcAMP had no apoptotic effect on the C6 glioma cells. There was an increase in the protein expression GFAP (specific marker for astrocytes). There was no significant effect between undifferentiated and 5-day differentiation regarding their response to glucose 10 mM. In addition, there were no significant effects of glucose on the basal of 5-day differentiation of C6 glioma cells. However, there was a significant correlation between the concentration of glucose and inhibition of the basal oxygen consumption. Finally, glucose 10 mM did not stimulate NAD (P)H levels of C6 glioma cells. CONCLUSION The above results showed that cAMP induce C6 glioma cells differentiation without affecting its bioenergetics. Therefore cAMP is considered to be the best differentiating agent.
Collapse
Affiliation(s)
- Ahmed S Hisab
- a School of life Sciences , Queens Medical Centre , Nottingham , UK.,b Department of internal medicine, Faculty of veterinary medicine , Basrah University , Basra , Iraq
| |
Collapse
|
11
|
Mijatović S, Bramanti A, Nicoletti F, Fagone P, Kaluđerović GN, Maksimović-Ivanić D. Naturally occurring compounds in differentiation based therapy of cancer. Biotechnol Adv 2018; 36:1622-1632. [DOI: 10.1016/j.biotechadv.2018.04.001] [Citation(s) in RCA: 24] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2017] [Revised: 03/22/2018] [Accepted: 04/10/2018] [Indexed: 12/22/2022]
|
12
|
Dibutyryl cAMP- or Interleukin-6-induced astrocytic differentiation enhances mannose binding lectin (MBL)-associated serine protease (MASP)-1/3 expression in C6 glioma cells. Arch Biochem Biophys 2018; 653:39-49. [PMID: 29963999 DOI: 10.1016/j.abb.2018.06.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2018] [Revised: 06/26/2018] [Accepted: 06/27/2018] [Indexed: 10/28/2022]
Abstract
Mannose-binding lectin (MBL)-Associated Serine Proteases (MASP)-1 and 3, key enzymes in the lectin complement pathway of innate immune response, are also expressed in glioma cell lines. We investigated MASP-1 and MASP-3 expression during dibutyryl cyclic AMP (dbcAMP)- or Interleukin-6 (rIL-6)-induced astrocytic differentiation of C6 glioma cells. Our results demonstrate that C6 cells express basal levels of MASP-1 and MASP-3 and following exposure to dbcAMP or IL-6, a consistent MASP-1 and MASP-3 mRNA up-regulation was found, with a behavior similar to that showed by the fibrillary acidic protein (GFAP). Furthermore, in cell conditioned media, rIL-6 stimulated MASP-3 secretion which reached levels similar to those obtained by dbcAMP treatment. Moreover, the detection of a 46-kDa MASP-3 suggested its processing to the mature form in the extracellular cell medium. Interestingly, the H89 PKA inhibitor, mostly affected dbcAMP-induced MASP-1 and MASP-3 mRNA levels, compared to that of rIL-6, suggesting that cAMP/PKA pathway contributes to MASP-1 and MASP-3 up-regulation. MASP-1 and MASP-3 expression increase was concomitant with dbcAMP- or rIL-6-induced phosphorylation of STAT3. Our findings suggest that the increase in intracellular cAMP concentration or rIL-6 stimulation can play a role in innate immunity enhancing MASP-1 and MASP-3 expression level in C6 glioma cells.
Collapse
|
13
|
Crespo MC, Tomé-Carneiro J, Pintado C, Dávalos A, Visioli F, Burgos-Ramos E. Hydroxytyrosol restores proper insulin signaling in an astrocytic model of Alzheimer's disease. Biofactors 2017; 43:540-548. [PMID: 28317262 DOI: 10.1002/biof.1356] [Citation(s) in RCA: 34] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2016] [Revised: 01/16/2017] [Accepted: 02/08/2017] [Indexed: 12/23/2022]
Abstract
Recent epidemiological evidence demonstrated that diabetes is a risk factor for AD onset and development. Indeed, meta-analyses of longitudinal epidemiologic studies show that diabetes increases AD risk by 50-100%, being insulin resistance (IR) the main binding link between diabetes and AD. Astrocytes are the foremost cerebral macroglial cells and are responsible for converting glucose into lactate and transfer it to neurons that use it as fuel, but Aβ(1-42) impairs insulin signaling and glycogen storage. Recent prospective studies showed that the Mediterranean diet is associated with lower incidence of AD. We hypothesized that hydroxytyrosol (HT, the preeminent polyphenol of olives and olive oil) could exert beneficial effects on IR associated with AD and investigated it mechanisms of action in an astrocytic model of AD. The astrocytic cell line C6 was exposed to Aβ(25-35) and co-incubated with HT for different periods. After treatment with Aβ(25-35), astrocytes' viability was significantly decreased as compared with controls; however, both pre- and post-treatment with HT prevented this effect. Mechanistically, we found that the preventive role of HT on Aβ(25-35)- induced cytotoxicity in astrocytes is moderated by an increased HT-induced activation of Akt, which is mediated by the insulin signaling pathway. In addition, we report that HT prevented the pronounced activation of mTOR, thereby restoring proper insulin signaling. In conclusion, we demonstrate that HT protects Aβ(25-35)-treated astrocytes by improving insulin sensitivity and restoring proper insulin-signaling. These data provide some mechanistic insight on the observed inverse association between olive oil consumption and prevalence of cognitive impairment. © 2017 BioFactors, 43(4):540-548, 2017.
Collapse
Affiliation(s)
| | | | - Cristina Pintado
- Facultad de Ciencias Ambientales y Bioquímica, Universidad de Castilla-La Mancha, Toledo, Spain
| | | | - Francesco Visioli
- IMDEA-Food, CEI UAM+CSIC, Madrid, Spain
- Department of Molecular Medicine, University of Padova, Padova, Italy
| | - Emma Burgos-Ramos
- Facultad de Ciencias Ambientales y Bioquímica, Universidad de Castilla-La Mancha, Toledo, Spain
| |
Collapse
|
14
|
Masutomi H, Kawashima S, Kondo Y, Uchida Y, Jang B, Choi EK, Kim YS, Shimokado K, Ishigami A. Induction of peptidylarginine deiminase 2 and 3 by dibutyryl cAMP via cAMP-PKA signaling in human astrocytoma U-251MG cells. J Neurosci Res 2016; 95:1503-1512. [PMID: 27704563 DOI: 10.1002/jnr.23959] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2016] [Revised: 08/30/2016] [Accepted: 09/12/2016] [Indexed: 12/16/2022]
Abstract
Peptidylarginine deiminases (PADs) are posttranslational modification enzymes that citrullinate (deiminate) protein arginine residues in a calcium-dependent manner, yielding citrulline residues. Enzymatic citrullination abolishes positive charges of native protein molecules, inevitably causing significant alterations in their structure and function. Previously, we reported the abnormal accumulation of citrullinated proteins and an increase of PAD2 content in hippocampi of patients with Alzheimer disease. In this study, we investigated PAD expression by using dibutyryl cAMP (dbcAMP) in human astrocytoma U-251MG cells. Under normal culture conditions, PAD2 and PAD3 mRNA expression is detectable with quantitative PCR in U-251MG cells. The addition of dbcAMP in a dose-dependent manner significantly increased this mRNA expression and protein levels. Moreover, PAD enzyme activity also increased significantly and dose-dependently. Furthermore, the expression of PAD2 and PAD3 mRNA was inhibited by the cAMP-dependent PKA inhibitor KT5720, suggesting that such expression of dbcAMP-induced PAD2 and PAD3 mRNA is mediated by the cAMP-PKA signaling pathway in U-251MG cells. This is the first report to document the PAD2 and PAD3 mRNA expression induced by dbcAMP and to attribute the induction of these genes to mediation by the cAMP-PKA signaling pathway in U-251MG cells. © 2016 Wiley Periodicals, Inc.
Collapse
Affiliation(s)
- Hirofumi Masutomi
- Molecular Regulation of Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan.,Geriatrics and Vascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Saki Kawashima
- Molecular Regulation of Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Yoshitaka Kondo
- Molecular Regulation of Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| | - Yoshiaki Uchida
- Research & Development Division, Fujirebio Inc., Tokyo, Japan
| | - Byungki Jang
- Ilsong Institute of Life Science, Hallym University, Anyang, Gyeonggi-do, Korea
| | - Eun-Kyoung Choi
- Ilsong Institute of Life Science, Hallym University, Anyang, Gyeonggi-do, Korea
| | - Yong-Sun Kim
- Ilsong Institute of Life Science, Hallym University, Anyang, Gyeonggi-do, Korea
| | - Kentaro Shimokado
- Geriatrics and Vascular Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | - Akihito Ishigami
- Molecular Regulation of Aging, Tokyo Metropolitan Institute of Gerontology, Tokyo, Japan
| |
Collapse
|
15
|
Leng Y, Wang J, Wang Z, Liao HM, Wei M, Leeds P, Chuang DM. Valproic Acid and Other HDAC Inhibitors Upregulate FGF21 Gene Expression and Promote Process Elongation in Glia by Inhibiting HDAC2 and 3. Int J Neuropsychopharmacol 2016; 19:pyw035. [PMID: 27207921 PMCID: PMC5006201 DOI: 10.1093/ijnp/pyw035] [Citation(s) in RCA: 29] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/07/2015] [Accepted: 04/18/2016] [Indexed: 12/13/2022] Open
Abstract
BACKGROUND Fibroblast growth factor 21, a novel regulator of glucose and lipid metabolism, has robust protective properties in neurons. However, its expression and function in glia are unknown. Valproic acid, a mood stabilizer and anticonvulsant, is a histone deacetylase inhibitor and a dynamic gene regulator. We investigated whether histone deacetylase inhibition by valproic acid and other inhibitors upregulates fibroblast growth factor 21 expression and, if so, sought to identify the histone deacetylase isoform(s) involved and their role in altering glial cell morphology. METHODS C6 glioma or primary cortical glial cultures were treated with histone deacetylase inhibitors, and fibroblast growth factor 21 levels and length of cell processes were subsequently measured. Histone deacetylase 1, 2, or 3 was also knocked down to detect which isoform was involved in regulating fibroblast growth factor 21 mRNA levels. Finally, knockdown and overexpression of fibroblast growth factor 21 were performed to determine whether it played a role in regulating cell process length. RESULTS Treatment of C6 cells or primary glial cultures with valproic acid elevated fibroblast growth factor 21 mRNA levels, extended cell process length, and markedly increased acetylated histone-H3 levels. Other histone deacetylase inhibitors including pan- and class I-specific inhibitors, or selective knockdown of histone deacetylase 2 or 3 isoform produced similar effects. Knockdown or overexpression of fibroblast growth factor 21 significantly decreased or increased C6 cell process length, respectively. CONCLUSIONS In glial cell line and primary glia, using pharmacological inhibition and selective gene silencing of histone deacetylases to boost fibroblast growth factor 21 mRNA levels results in elongation of cell processes. Our study provides a new mechanism via which histone deacetylase 2 and 3 participate in upregulating fibroblast growth factor 21 transcription and extending process outgrowth in glia.
Collapse
Affiliation(s)
- Yan Leng
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, Bethesda, MD.
| | | | | | | | | | | | - De-Maw Chuang
- Molecular Neurobiology Section, National Institute of Mental Health, National Institutes of Health, Bethesda, MD.
| |
Collapse
|
16
|
McCarthy RC, Lu DY, Alkhateeb A, Gardeck AM, Lee CH, Wessling-Resnick M. Characterization of a novel adult murine immortalized microglial cell line and its activation by amyloid-beta. J Neuroinflammation 2016; 13:21. [PMID: 26819091 PMCID: PMC4730646 DOI: 10.1186/s12974-016-0484-z] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2015] [Accepted: 01/18/2016] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND Alzheimer's disease is associated with amyloid-beta (Aβ)-induced microglia activation. This pro-inflammatory response promotes neuronal damage, and therapies are sought to limit microglial activation. Screening efforts to develop new pharmacological inhibitors require a robust in vitro cell system. Current models lack significant responses to Aβ, and their use in examining age-related neurodegenerative diseases is questionable. For example, the commonly used BV-2 microglial line was derived from embryonic mononuclear cells and its activation by various stimuli is limited. To this end, we have established a new immortalized microglial (IMG) cell line from adult murine brain. The objective of this study was to characterize Aβ-induced activation of IMG cells, and here, we demonstrate the ability of cannabinoids to significantly reduce this inflammatory response. METHODS Microglial cells derived from adult murine brain were immortalized via infection with the v-raf/v-myc retrovirus under conditions that selectively promote microglia growth. The presence or absence of markers CD11b and F4/80 (microglial), NeuN (neuronal), and GFAP (astrocytic) was assessed by immunofluorescence microscopy and western blotting. Using IMG and BV-2 cells, levels of pro- and anti-inflammatory transcripts in response to extracellular stimuli were determined by quantitative PCR (qPCR). Phagocytosis of fluorescent beads and fluorescein isothiocyanate (FITC)-labeled Aβ oligomers was assessed using flow cytometry and fluorescence microscopy. FITC-Aβ uptake was quantified using a fluorescence plate reader. The ability of cannabinoids to mitigate Aβ-induced expression of inducible nitric oxide synthase (iNOS) was evaluated. RESULTS IMG cells express the microglial markers CD11b and F4/80 but not NeuN or GFAP. Relative to BV-2 cells, IMG cells increased iNOS (>200-fold) and Arg-1 (>100-fold) in response to pro- and anti-inflammatory stimuli. IMG cells phagocytose foreign particles and Aβ oligomers, with the latter trafficked to phagolysosomes. Aβ-induced activation of IMG cells was suppressed by delta-9-tetrahydrocannabinol and the CB2-selective agonist JWH-015 in a time- and concentration-dependent manner. CONCLUSIONS IMG cells recapitulate key features of microglial cell activation. As an example of their potential pharmacological use, cannabinoids were shown to reduce activation of Aβ-induced iNOS gene expression. IMG cells hold promising potential for drug screening, mechanistic studies, and functional investigations directed towards understanding how Aβ interacts with microglia.
Collapse
Affiliation(s)
- Ryan C McCarthy
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, USA.
| | - Dah-Yuu Lu
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, USA.
- Present Address: Graduate Institute of Neural and Cognitive Sciences, China Medical University, Taichung, Taiwan, Republic of China.
| | - Ahmed Alkhateeb
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, USA.
- Present Address: The Wellman Center for Photomedicine, Massachusetts General Hospital and Harvard Medical School, Boston, MA, USA.
| | - Andrew M Gardeck
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, USA.
| | - Chih-Hao Lee
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, USA.
| | - Marianne Wessling-Resnick
- Department of Genetics and Complex Diseases, Harvard School of Public Health, 655 Huntington Avenue, Boston, MA, 02115, USA.
| |
Collapse
|
17
|
Chao CC, Kan D, Lo TH, Lu KS, Chien CL. Induction of neural differentiation in rat C6 glioma cells with taxol. Brain Behav 2015; 5:e00414. [PMID: 26665000 PMCID: PMC4667627 DOI: 10.1002/brb3.414] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Revised: 09/14/2015] [Accepted: 09/21/2015] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Glioblastoma is a common and aggressive type of primary brain tumor. Several anticancer drugs affect GBM (glioblastoma multiforme) cells on cell growth and morphology. Taxol is one of the widely used antineoplastic drugs against many types of solid tumors, such as breast, ovarian, and prostate cancers. However, the effect of taxol on GBM cells remains unclear and requires further investigation. METHODS Survival rate of C6 glioma cells under different taxol concentrations was quantified. To clarify the differentiation patterns of rat C6 glioma cells under taxol challenge, survived glioma cells were characterized by immunocytochemical, molecular biological, and cell biological approaches. RESULTS After taxol treatment, not only cell death but also morphological changes, including cell elongation, cellular processes thinning, irregular shapes, and fragmented nucleation or micronuclei, occurred in the survived C6 cells. Neural differentiation markers NFL (for neurons), β III-tubulin (for neurons), GFAP (for astrocytes), and CNPase (for oligodendrocytes) were detected in the taxol-treated C6 cells. Quantitative analysis suggested a significant increase in the percentage of neural differentiated cells. The results exhibited that taxol may trigger neural differentiation in C6 glioma cells. Increased expression of neural differentiation markers in C6 cells after taxol treatment suggest that some anticancer drugs could be applied to elimination of the malignant cancer cells as well as changing proliferation and differentiation status of tumor cells.
Collapse
Affiliation(s)
- Chuan-Chuan Chao
- Department of Anatomy and Cell Biology College of Medicine National Taiwan University Taipei Taiwan
| | - Daphne Kan
- Center of Genomic Medicine National Taiwan University Taipei Taiwan
| | - Ta-Hsuan Lo
- Center of Genomic Medicine National Taiwan University Taipei Taiwan
| | - Kuo-Shyan Lu
- Department of Anatomy and Cell Biology College of Medicine National Taiwan University Taipei Taiwan
| | - Chung-Liang Chien
- Department of Anatomy and Cell Biology College of Medicine National Taiwan University Taipei Taiwan; Center of Genomic Medicine National Taiwan University Taipei Taiwan
| |
Collapse
|
18
|
Muniroh M, Khan N, Koriyama C, Akiba S, Vogel CFA, Yamamoto M. Suppression of methylmercury-induced IL-6 and MCP-1 expressions by N-acetylcysteine in U-87MG human astrocytoma cells. Life Sci 2015; 134:16-21. [PMID: 26006043 DOI: 10.1016/j.lfs.2015.04.024] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2015] [Revised: 04/09/2015] [Accepted: 04/23/2015] [Indexed: 10/23/2022]
Abstract
AIMS The aim of this study was to clarify the involvement of oxidative stress in methylmercury (MeHg)-induced pro-inflammatory cytokine expressions and the suppressive effects of N-acetylcysteine (NAC) in MeHg-induced cytokine expression. MATERIALS AND METHODS Using U-87-MG human astrocytoma cell line, interleukin (IL)-6 and monocyte chemoattractant protein (MCP)-1 expressions induced by 4 μM MeHg were measured at mRNA and protein levels. Hydrogen peroxide (H2O2) and superoxide anion (O2(-)) were quantified by flow-cytometry analysis. To examine the suppressive effects of NAC on the cytokine expressions among different timing of NAC treatment, cells were treated with 0.5 or 5mM NAC before, simultaneously, or after MeHg administration. KEY FINDINGS MeHg exposure at 4 μM, a non-cytotoxic concentration, significantly induced MCP-1 and IL-6 expressions at both mRNA and protein levels. A significant increase of H2O2 production but not O2(-) was observed. MeHg-induced expression of MCP-1 and IL-6 mRNA was reduced by 10-20% in the presence of 5mM NAC (co-treatment experiment) compared to cells treated with MeHg only. Pre-treatment of cells with 0.5 or 5mM NAC at 0.5 or 1h and its subsequent washout before MeHg addition suppressed MCP-1 and IL-6 cytokine expressions. Post-treatment of cells with NAC after MeHg addition also suppressed the cytokine induction, but the magnitude of suppression was evidently lower than in co-treated cells even though the H2O2 generation was almost completely suppressed by NAC. SIGNIFICANCE NAC may effectively suppress the MeHg-induced cytokine production through both, inhibition of reactive oxygen species as well as extracellular chelation of MeHg in astrocytes.
Collapse
Affiliation(s)
- Muflihatul Muniroh
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan; Department of Physiology, Faculty of Medicine, Diponegoro University, Tembalang, Semarang 50725, Indonesia
| | - Noureen Khan
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | - Chihaya Koriyama
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan.
| | - Suminori Akiba
- Department of Epidemiology and Preventive Medicine, Kagoshima University Graduate School of Medical and Dental Sciences, 8-35-1 Sakuragaoka, Kagoshima 890-8520, Japan
| | - Christoph F A Vogel
- Department of Environmental Toxicology and Center for Health and the Environment, University of California, Davis, CA 95616, USA
| | - Megumi Yamamoto
- Integrated Physiology Section, Department of Basic Medical Science, National Institute for Minamata Disease, 4058-18 Hama, Minamata, Kumamoto 867-0008, Japan
| |
Collapse
|
19
|
Chao CC, Kan D, Lu KS, Chien CL. The role of microRNA-30c in the self-renewal and differentiation of C6 glioma cells. Stem Cell Res 2015; 14:211-23. [PMID: 25698399 DOI: 10.1016/j.scr.2015.01.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/22/2014] [Revised: 01/04/2015] [Accepted: 01/26/2015] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Sphere formation, one method for identifying self-renewal ability, has been used to report that cancer stem-like cells exist in rat C6 glioma cells. Recent studies suggested that cancer stem-like cells share the stem cell properties of self-renewal and multipotent ability of neural stem cells and might be regulated by microRNAs (miRNAs). However, the mechanism of miRNA involvement in the sphere formation and neural differentiation abilities of cancer stem-like cells is poorly understood. RESULTS We found that miRNA-30c could assist in sphere formation of C6 cells under defined conditions in neural stem cell medium DMEM/F12-bFGF-EGF-B27. Moreover, overexpression of miRNA-30c might reduce 3-isobutyl-1-methylxanthine (IBMX)-induced neural differentiation, as the expression of neural markers, especially glial fibrillary acidic protein (GFAP), decreased. Further experiments revealed that miRNA-30c inhibited the IBMX-induced astrocyte differentiation via targeting the upstream genes and inactivating phosphorylation of STAT3 of the JAK-STAT3 pathway. Subsequently, the expression of GFAP was reduced and the number of astrocyte differentiation from C6 cells decreased. CONCLUSIONS Our findings suggest that miRNA-30c could play a regulatory role in self-renewal and neural differentiation in C6 glioma cells.
Collapse
Affiliation(s)
- Chuan-Chuan Chao
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Daphne Kan
- Center of Genomic Medicine, National Taiwan University, 6F., No. 2, Syu-Jhou Road, Taipei 100, Taiwan
| | - Kuo-Shyan Lu
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei 100, Taiwan
| | - Chung-Liang Chien
- Department of Anatomy and Cell Biology, College of Medicine, National Taiwan University, No. 1, Section 1, Jen-Ai Road, Taipei 100, Taiwan; Center of Genomic Medicine, National Taiwan University, 6F., No. 2, Syu-Jhou Road, Taipei 100, Taiwan.
| |
Collapse
|
20
|
Barbosa DJ, Capela JP, de Lourdes Bastos M, Carvalho F. In vitro models for neurotoxicology research. Toxicol Res (Camb) 2015; 4:801-842. [DOI: 10.1039/c4tx00043a] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 09/01/2023] Open
Abstract
The nervous system has a highly complex organization, including many cell types with multiple functions, with an intricate anatomy and unique structural and functional characteristics; the study of its (dys)functionality following exposure to xenobiotics, neurotoxicology, constitutes an important issue in neurosciences.
Collapse
Affiliation(s)
- Daniel José Barbosa
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| | - João Paulo Capela
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| | - Maria de Lourdes Bastos
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| | - Félix Carvalho
- REQUIMTE (Rede de Química e Tecnologia)
- Laboratório de Toxicologia
- Departamento de Ciências Biológicas
- Faculdade de Farmácia
- Universidade do Porto
| |
Collapse
|
21
|
Zhou W, Jiang Z, Li X, Xu Y, Shao Z. Cytokines: shifting the balance between glioma cells and tumor microenvironment after irradiation. J Cancer Res Clin Oncol 2014; 141:575-89. [PMID: 25005789 DOI: 10.1007/s00432-014-1772-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2014] [Accepted: 06/30/2014] [Indexed: 12/13/2022]
Abstract
Malignant gliomas invariably recur after irradiation, showing radioresistance. Meanwhile, cranial irradiation can bring some risk for developing cognitive dysfunction. There is increasing evidence that cytokines play their peculiar roles in these processes. On the one hand, cytokines directly influence the progression of malignant glioma, promoting or suppressing tumor progression. On the other hand, cytokines indirectly contribute to the immunologic response against gliomas, exhibiting pro-inflammatory or immunosuppressive activities. We propose that cytokines are not simply unregulated products from tumor cells or immune cells, but mediators finely adjust the balance between glioma cells and tumor microenvironment after irradiation. The paper, therefore, focuses on the changes of cytokines after irradiation, analyzing how these mediate the response of tumor cells and normal cells to irradiation. In addition, cytokine-based immunotherapeutic strategies, accompanied with irradiation, for the treatment of gliomas are also discussed.
Collapse
Affiliation(s)
- Wei Zhou
- Department of Radiation Oncology, Cancer Centre, Qilu Hospital, Shandong University, 44 Wenhuaxi Road, Jinan, 250012, Shandong, China
| | | | | | | | | |
Collapse
|
22
|
Involvement of the Notch pathway in terminal astrocytic differentiation: role of PKA. ASN Neuro 2013; 5:e00130. [PMID: 24286475 PMCID: PMC3891361 DOI: 10.1042/an20130023] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
The Notch pathway is a highly conserved signaling system essential for modulating neurogenesis and promoting astrogenesis. Similarly, the cAMP signaling cascade can promote astrocytic commitment in several cell culture models, such as the C6 glioma cell line. These cells have the capacity to differentiate into oligodendrocytes or astrocytes, characteristics that allow their use as a glial progenitor model. In this context, we explore here the plausible involvement of cAMP in Notch-dependent signal transactions. The exposure of C6 cells to a non-hydrolysable cAMP analogue resulted in a sustained augmentation of Notch activity, as detected by nuclear translocation of its intracellular domain portion (NICD) and transcriptional activity. The cAMP effect is mediated through the activation of the γ-secretase complex, responsible for Notch cleavage and is sensitive to inhibitors of the cAMP-dependent protein kinase, PKA. As expected, Notch cleavage and nuclear translocation resulted in the up-regulation of the mRNA levels of one of its target genes, the transcription factor Hair and enhancer of split 5. Moreover, the glutamate uptake activity, as well as the expression of astrocytic markers such as glial fibrillary acidic protein, S100β protein and GLAST was also enhanced in cAMP-exposed cells. Our results clearly suggest that during the process of C6 astrocytic differentiation, cAMP activates the PKA/γ-secretase/NICD/RBPJκ pathway and Notch1 expression, leading to transcriptional activation of the genes responsible for glial progenitor cell fate decision.
Collapse
|
23
|
Adornetto A, Pagliara V, Renzo GD, Arcone R. Polychlorinated biphenyls impair dibutyryl cAMP-induced astrocytic differentiation in rat C6 glial cell line. FEBS Open Bio 2013; 3:459-66. [PMID: 24251112 PMCID: PMC3829991 DOI: 10.1016/j.fob.2013.10.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/11/2013] [Revised: 10/20/2013] [Accepted: 10/22/2013] [Indexed: 02/04/2023] Open
Abstract
In the central nervous system, alteration of glial cell differentiation can affect brain functions. Polychlorinated biphenyls (PCBs) are persistent environmental chemical contaminants that exert neurotoxic effects in glial and neuronal cells. We examined the effects of a commercial mixture of PCBs, Aroclor1254 (A1254) on astrocytic differentiation of glial cells, using the rat C6 cell line as in vitro model. The exposure for 24 h to sub-toxic concentrations of A1254 (3 or 9 μM) impaired dibutyryl cAMP-induced astrocytic differentiation as showed by the decrease of glial fibrillary acidic protein (GFAP) protein levels and inhibition in change of cell morphology toward an astrocytic phenotype. The A1254 inhibition was restored by the addition of a protein kinase C (PKC) inhibitor, bisindolylmaleimide (bis), therefore indicating that PCBs disturbed the cAMP-induced astrocytic differentiation of C6 cells via the PKC pathway. The phosphorylation of signal transducer and activator of transcription 3 (STAT3) is essential for cAMP-induced transcription of GFAP promoter in C6 cells. Our results indicated that the exposure to A1254 (3 or 9 μM) for 24 h suppressed cAMP-induced STAT3 phosphorylation. Moreover, A1254 reduced cAMP-dependent phosphorylation of STAT3 requires inhibition of PKC activity. Together, our results suggest that PCBs induce perturbation in cAMP/PKA and PKC signaling pathway during astrocytic differentiation of glial cells.
Collapse
Key Words
- A1254, Aroclor 1254
- Aroclor1254
- Astrocytic differentiation
- C6 glial cell line
- CNS, central nervous system
- CRE, cAMP responsive element
- CREB, cAMP-response element binding protein
- DAPI, 4′,6-diamidino-2-phenylindole
- DMEM, Dulbecco’s Modified Eagle’s Medium
- DMSO, dimethyl sulfoxide
- GAPDH, glyceraldehyde-3-phosphate dehydrogenase
- GFAP, glial fibrillary acidic protein
- Glial fibrillary acidic protein (GFAP)
- MTT, 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl-2H-tetrazolium bromide
- NMDA, N-methyl-d-aspartate
- PCBs, polychlorinated biphenyls
- PKA, protein kinase A
- PKC, protein kinase C
- Protein kinase C (PKC)
- ROS, reactive oxygen species
- STAT3, signal transducer and activator of transcription 3
- Signal transducer and activator of transcription 3 (STAT3)
- TRE, CRE transcriptional response element
- bis, 2-[1-(3-dimethylamino-propyl)indol-3-yl]-3-(indol-3-yl) maleimide
- dbcAMP, N6,2′-O-dibutyryl cAMP
- nNOS, neuronal nitric oxide
Collapse
Affiliation(s)
- Annagrazia Adornetto
- Department of Pharmacy, Health and Nutritional Sciences, University of Calabria, Via P. Bucci, Arcavacata di Rende, Cosenza (CS) 87036, Italy
| | | | | | | |
Collapse
|
24
|
Acetylcholine esterase is a regulator of GFAP expression and a target of dichlorvos in astrocytic differentiation of rat glioma C6 cells. Brain Res 2013; 1537:37-45. [PMID: 24001591 DOI: 10.1016/j.brainres.2013.08.031] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2013] [Accepted: 08/16/2013] [Indexed: 02/07/2023]
Abstract
The main target of neurotoxins is neurons because they comprise the main part of neural function, but glial cells may be indirect targets because they support the function of neurons. Among the glial cells, astrocytes in particular act as "nurse cells", regulating neuronal survival and functions. In the present study, to reveal whether a known neurotoxic substance, organophosphate dichlorvos (DDVP), affects the differentiation of astrocytes, we used an astrocyte differentiation model in rat glioma C6 cells. Morphological change and induction of GFAP expression in the differentiating C6 cells were suppressed by DDVP treatment. The known potential targets of DDVP are acetylcholine esterase (AChE), fatty acid amide hydrolase and methyl guanine methyl transferase. Among the specific inhibitors against these enzymes, the AChE inhibitor paraoxon successfully suppressed the cellular morphological changes and the induction of GFAP expression in differentiating C6 cells. These results indicate that DDVP inhibits differentiation in the C6 astrocyte-differentiation model, in which at least AChE inhibition is involved and that AChE is a potent regulator of the differentiation. Furthermore, considering that the main substrate of AChE is ACh, thus, ACh may act as regulators of astrocyte differentiation.
Collapse
|
25
|
Yamada T, Urano-Tashiro Y, Hashi Y, Sakumoto M, Akiyama H, Tashiro F. The U-box-type ubiquitin ligase PRP19β regulates astrocyte differentiation via ubiquitination of PTP1B. Brain Res 2013; 1524:12-25. [DOI: 10.1016/j.brainres.2013.06.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/18/2012] [Revised: 06/01/2013] [Accepted: 06/05/2013] [Indexed: 11/15/2022]
|
26
|
Hai J, Lin Q, Lu Y, Yi J, Zhang H. Growth inhibition and induction of differentiation by panaxydol in rat C6 glioma cells. Neurol Res 2013; 30:99-105. [PMID: 17767808 DOI: 10.1179/016164107x228697] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES Panaxydol is a naturally occurring non-peptidyl small molecule isolated from the lipophilic fractions of Panax notoginseng, a well-known Chinese traditional medicine. In this study, we aimed to investigate the effects of panaxydol on growth inhibition and its mechanisms in C6 rat glioma cells. METHODS The effects of panaxydol on cell proliferation, morphologic changes, glial fibrillary acidic protein (GFAP) expression and cell cycle regulation in rat C6 cells were evaluated using 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyl tetrazolium bromide (MTT) assay, hematoxylin and eosin (HE) staining, immunocytochemistry, flow cytometric analysis and Western blot respectively. RESULTS Panaxydol markedly inhibited the proliferation of C6 cells in a dose-dependent manner with IC50 of 39.5 +/- 2.3 microM. In addition, the cell morphologic changes and increased expression of GFAP in C6 cells in the presence of panaxydol implied a cellular differentiation. Flow cytometric analysis revealed that panaxydol-treated cells accumulated in G0/G1 phase with a marked decrease in the number of C6 cells at S phase. Western blot analysis demonstrated that panaxydol resulted in an increase in the protein expression of p27 in C6 cells as early as 3 hours after treatment consistent with the differentiation response, but protein expression of p53, p21, p16 and pRb remained unchanged. CONCLUSION These findings suggest that panaxydol inhibits the proliferation of C6 cells via G0/G1 cell cycle arrest in association with induction of p27 expression and differentiation.
Collapse
Affiliation(s)
- Jian Hai
- Department of Neurosurgery, Tongji Hospital, Tongji University, Shanghai, China.
| | | | | | | | | |
Collapse
|
27
|
de Souza DF, Wartchow K, Hansen F, Lunardi P, Guerra MC, Nardin P, Gonçalves CA. Interleukin-6-induced S100B secretion is inhibited by haloperidol and risperidone. Prog Neuropsychopharmacol Biol Psychiatry 2013; 43:14-22. [PMID: 23246638 DOI: 10.1016/j.pnpbp.2012.12.001] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Revised: 11/21/2012] [Accepted: 12/04/2012] [Indexed: 01/03/2023]
Abstract
Although inflammation may be a physiological defense process, imbalanced neuroinflammation has been associated with the pathophysiology of brain disorders, including major depression and schizophrenia. Activated glia releases a variety of pro-inflammatory cytokines that contribute to neuronal dysfunction. Elevated levels of S100B, a glia derived protein, have been observed in the serum and CSF of schizophrenic patients suggesting a glial role in the disease. We evaluated whether S100B secretion (in C6 glioma cells and hippocampal slices in Wistar rats) could be directly modulated by the main inflammatory cytokines (IL-1β, TNF-α, IL-6 and IL-8) altered in schizophrenia, as well as the possible involvement of mitogen-activated protein kinase (MAPK) pathways in these responses. We also investigated the effects of typical and atypical antipsychotic drugs on glial cytokine-induced S100B release. Our results suggest that S100B secretion is increased by pro-inflammatory cytokines via MAPK and that oxidative stress may be a component of this modulation. These results reinforce the idea that the S100B protein is involved in the inflammatory response observed in many brain diseases, including schizophrenia. Moreover the antipsychotics, haloperidol and risperidone, were able to inhibit the secretion of S100B following IL-6 stimulation in C6 glioma cells.
Collapse
Affiliation(s)
- Daniela Fraga de Souza
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Ramiro Barcelos, 2600-Anexo, 90035-003, Porto Alegre, Brazil
| | | | | | | | | | | | | |
Collapse
|
28
|
Characterization of the expression and cell-surface localization of transmembrane protein 132A. Mol Cell Biochem 2012; 370:23-33. [PMID: 22821197 DOI: 10.1007/s11010-012-1394-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/02/2012] [Accepted: 07/07/2012] [Indexed: 10/28/2022]
Abstract
Transmembrane protein 132A (TMEM132A, KIAA1583) was first isolated as a novel gene that is enriched during the embryonic and postnatal stages of rat brain development and interacts with GRP78. However, the biological functions of TMEM132A are scarcely characterized because the protein does not contain any known structural domains. Using a cell-surface biotinylation assay and immunocytochemical staining, we found that TMEM132A is a transmembrane glycoprotein consisting of a large extracellular domain in the N-terminal region and a small cytosolic domain in the C-terminal region. Partial deletions of the intracellular domain of TMEM132A had little effect on its expression level and cell-surface localization in transfected HEK293 cells, whereas deletions of the extracellular domain hampered transport to the cell surface. The expression pattern of each N-terminal mutant was immunocytochemically confirmed in HeLa cells transfected with the same constructs. Treatment with tunicamycin, an inhibitor of protein glycosylation, led to the accumulation of the unglycosylated form of TMEM132A in inverse proportion to the glycosylated form; however, both forms were localized at the cell surface at almost equal rates. In contrast, GRP78 overexpression led to the accumulation of unglycosylated TMEM132A, which was not detected on the cell surface. Inhibition of ER-Golgi transport by treatment with brefeldin A or the overexpression of mutant Sar1 attenuated the amount of cell-surface localized TMEM132A in HEK293 cells. Treatment with reagents disrupting intracellular calcium rapidly down-regulated the amount of TMEM132A protein in Neuro2a cells without affecting the expression level of its mRNA. Taken together, our data show that the novel cell-surface localized glycoprotein, TMEM132A, is regulated by several factors, including GRP78, Sar1, and intracellular calcium, in a post-transcriptional manner.
Collapse
|
29
|
Shu M, Zhou Y, Zhu W, Zhang H, Wu S, Chen J, Yan G. MicroRNA 335 is required for differentiation of malignant glioma cells induced by activation of cAMP/protein kinase A pathway. Mol Pharmacol 2011; 81:292-8. [PMID: 22172575 DOI: 10.1124/mol.111.076166] [Citation(s) in RCA: 45] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Glioma is the most common malignant cancer affecting the central nerve system, with dismal prognosis. Differentiation-inducing therapy is a novel strategy that has been preliminarily proved effective against malignant glioma. We have reported previously that activation of cAMP/protein kinase A (PKA) pathway is capable of inducing glioma cell differentiation, characterized by astrocyte-like shape and dramatic induction of astrocyte biomarker glial fibrillary acidic protein (GFAP). However, little progress has been made on molecular mechanisms related. Here we demonstrate that microRNA 335 (miR-335) is responsible for the glioma cell differentiation stimulated by activation of cAMP/PKA pathway. In the cAMP elevator cholera toxin-induced differentiation model of rat C6 glioma cells, miR-335 was significantly up-regulated, which was mimicked by other typical cAMP/PKA pathway activators (e.g., forskolin, dibutyryl-cAMP) and abolished by PKA-specific inhibitor (9R,10S,12S)-2,3,9,10,11,12-hexahydro-10-hydroxy-9-methyl-1-oxo-9,12-epoxy-1H-diindolo[1,2,3-fg:3',2',1'-kl]pyrrolo[3,4-i] [1,6]benzodiazocine-10-carboxylic acid, hexyl ester (KT5720). In an assay measuring gain and loss of miR-335 function, exogenetic miR-335 resulted in induction of GFAP, whereas miR-335 specific inhibitor antagomir-335 violently blocked cholera toxin-induced GFAP up-regulation. It is noteworthy that in human U87-MG glioma cells and human primary culture glioma cells, miR-335 also mediated cholera toxin-induced differentiation. Taken together, our findings suggest that miR-335 is potently required for differentiation of malignant glioma cells induced by cAMP/PKA pathway activation, and a single microRNA may act as an important fate determinant to control the differentiation status of malignant gliomas, which has provided a new insight into differentiation-inducing therapy against malignant gliomas.
Collapse
Affiliation(s)
- Minfeng Shu
- Department of Pharmacology, Zhongshan School of Medicine, Sun Yat-sen University, Guangzhou, PR China
| | | | | | | | | | | | | |
Collapse
|
30
|
Lee S, Kim JH, Kim JH, Seo JW, Han HS, Lee WH, Mori K, Nakao K, Barasch J, Suk K. Lipocalin-2 Is a chemokine inducer in the central nervous system: role of chemokine ligand 10 (CXCL10) in lipocalin-2-induced cell migration. J Biol Chem 2011; 286:43855-43870. [PMID: 22030398 DOI: 10.1074/jbc.m111.299248] [Citation(s) in RCA: 124] [Impact Index Per Article: 9.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The secreted protein lipocalin-2 (LCN2) has been implicated in diverse cellular processes, including cell morphology and migration. Little is known, however, about the role of LCN2 in the CNS. Here, we show that LCN2 promotes cell migration through up-regulation of chemokines in brain. Studies using cultured glial cells, microvascular endothelial cells, and neuronal cells suggest that LCN2 may act as a chemokine inducer on the multiple cell types in the CNS. In particular, up-regulation of CXCL10 by JAK2/STAT3 and IKK/NF-κB pathways in astrocytes played a pivotal role in LCN2-induced cell migration. The cell migration-promoting activity of LCN2 in the CNS was verified in vivo using mouse models. The expression of LCN2 was notably increased in brain following LPS injection or focal injury. Mice lacking LCN2 showed the impaired migration of astrocytes to injury sites with a reduced CXCL10 expression in the neuroinflammation or injury models. Thus, the LCN2 proteins, secreted under inflammatory conditions, may amplify neuroinflammation by inducing CNS cells to secrete chemokines such as CXCL10, which recruit additional inflammatory cells.
Collapse
Affiliation(s)
- Shinrye Lee
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Jong-Heon Kim
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Jae-Hong Kim
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Jung-Wan Seo
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Hyung-Soo Han
- Department of Physiology, Brain Science & Engineering Institute, Cell and Matrix Research Institute, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Won-Ha Lee
- Departments of School of Life Sciences and Biotechnology, Kyungpook National University School of Medicine, Daegu 700-422, Korea
| | - Kiyoshi Mori
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Kazuwa Nakao
- Department of Medicine and Clinical Science, Kyoto University Graduate School of Medicine, Kyoto 606-8507, Japan
| | - Jonathan Barasch
- Department of Medicine, College of Physicians and Surgeons, Columbia University, New York, New York 10027
| | - Kyoungho Suk
- Department of Pharmacology, Kyungpook National University School of Medicine, Daegu 700-422, Korea.
| |
Collapse
|
31
|
Sugimoto N, Miwa S, Ohno-Shosaku T, Tsuchiya H, Hitomi Y, Nakamura H, Tomita K, Yachie A, Koizumi S. Activation of tumor suppressor protein PTEN and induction of apoptosis are involved in cAMP-mediated inhibition of cell number in B92 glial cells. Neurosci Lett 2011; 497:55-9. [PMID: 21529704 DOI: 10.1016/j.neulet.2011.04.028] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2011] [Revised: 04/05/2011] [Accepted: 04/12/2011] [Indexed: 01/08/2023]
Abstract
During brain development, cAMP induces morphological changes and inhibits growth effects in several cell types. However, the molecular mechanisms underlying the growth inhibition remain unknown. Tumor suppressor protein phosphatase and tensin homolog deleted on chromosome 10 (PTEN) is a lipid phosphatase that inhibits the phosphoinositide 3-kinase (PI3K) pathway. The phosphorylation of Akt, which is one of the key molecules downstream of PI3K, inhibits apoptosis. In this study, we investigated the role of PTEN in cAMP-mediated growth inhibition. B92 rat glial cells were treated with 2 different cAMP stimulatory agents, a phosphodiesterase (PDE) inhibitor and a β-adrenoceptor agonist. Both cAMP stimulatory agents induced marked morphological changes in the cells, decreased cell number, decreased Akt phosphorylation, activated PTEN, cleaved caspase-3, and induced the condensation and fragmentation of nuclei. These results indicate that the cAMP stimulatory agents induced apoptosis. Protein phosphatase inhibitor prevented cAMP-induced dephosphorylation of PTEN and Akt. In addition, cAMP analogs and Epac-selective agonists affected PTEN and Akt activities. These results suggested that cAMP-induced apoptosis may be mediated by PTEN activation and Akt inhibition through protein phosphatase in B92 cells. Our results provide new insight into the role of PTEN in cAMP-induced apoptosis in glial cells.
Collapse
Affiliation(s)
- Naotoshi Sugimoto
- Department of Physiology, Graduate School of Medical Science, Kanazawa University, 13-1 Takara-machi, Ishikawa, Kanazawa 920-8640, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Liu X, Yang JM, Zhang SS, Liu XY, Liu DX. Induction of cell cycle arrest at G1 and S phases and cAMP-dependent differentiation in C6 glioma by low concentration of cycloheximide. BMC Cancer 2010; 10:684. [PMID: 21159181 PMCID: PMC3009684 DOI: 10.1186/1471-2407-10-684] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2010] [Accepted: 12/15/2010] [Indexed: 01/09/2023] Open
Abstract
Background Differentiation therapy has been shown effective in treatment of several types of cancer cells and may prove to be effective in treatment of glioblastoma multiforme, the most common and most aggressive primary brain tumor. Although extensively used as a reagent to inhibit protein synthesis in mammalian cells, whether cycloheximide treatment leads to glioma cell differentiation has not been reported. Methods C6 glioma cell was treated with or without cycloheximide at low concentrations (0.5-1 μg/ml) for 1, 2 and 3 days. Cell proliferation rate was assessed by direct cell counting and colony formation assays. Apoptosis was assessed by Hoechst 33258 staining and FACS analysis. Changes in several cell cycle regulators such as Cyclins D1 and E, PCNA and Ki67, and several apoptosis-related regulators such as p53, p-JNK, p-AKT, and PARP were determined by Western blot analysis. C6 glioma differentiation was determined by morphological characterization, immunostaining and Western blot analysis on upregulation of GFAP and o p-STAT3 expression, and upregulation of intracellular cAMP. Results Treatment of C6 cell with low concentration of cycloheximide inhibited cell proliferation and depleted cells at both G2 and M phases, suggesting blockade at G1 and S phases. While no cell death was observed, cells underwent profound morphological transformation that indicated cell differentiation. Western blotting and immunostaining analyses further indicated that changes in expression of several cell cycle regulators and the differentiation marker GFAP were accompanied with cycloheximide-induced cell cycle arrest and cell differentiation. Increase in intracellular cAMP, a known promoter for C6 cell differentiation, was found to be elevated and required for cycloheximide-promoted C6 cell differentiation. Conclusion Our results suggest that partial inhibition of protein synthesis in C6 glioma by low concentration of cycloheximide induces cell cycle arrest at G1 and M phases and cAMP-dependent cell differentiation.
Collapse
Affiliation(s)
- Xijun Liu
- Department of Neural and Behavioral Sciences, Penn State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | | | | | | | |
Collapse
|
33
|
Kulikov AV, Sinyakova NA, Naumenko VS, Bazovkina DV, Popova NK. Association of glycoprotein gp130 with hereditary catalepsy in mice. GENES BRAIN AND BEHAVIOR 2010; 9:997-1003. [DOI: 10.1111/j.1601-183x.2010.00639.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
|
34
|
Kulikov AV, Naumenko VS, Tsybko AS, Sinyakova NA, Bazovkina DV, Popova NK. The role of the glycoprotein gp130 in the serotonin mediator system in the mouse brain. Mol Biol 2010. [DOI: 10.1134/s0026893310050171] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
35
|
Lu WJ, Lan F, He Q, Lee A, Tang CZ, Dong L, Lan B, Ma X, Wu JC, Shen L. Inducible expression of stem cell associated intermediate filament nestin reveals an important role in glioblastoma carcinogenesis. Int J Cancer 2010; 128:343-51. [PMID: 20669222 DOI: 10.1002/ijc.25586] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2010] [Accepted: 07/20/2010] [Indexed: 12/17/2022]
Abstract
The intermediate filament nestin is transiently expressed in neural stem/progenitor cells during the development of central nervous system. Recently, increasing evidence has shown that upregulation of nestin is related to malignancy of several cancers, especially glioblastoma. However, the function of nestin in carcinogenesis remains unclear. In this study, we investigated the role of nestin in glioblastoma carcinogenesis by comparing subclones of rat C6 glioblastoma cells that were either high or low for nestin expression. We found that while nestin expression did not influence the in vitro proliferation of glioblastoma cells, subclones characterized by high levels of nestin formed tumors in vivo at significantly faster rates than subclones with low expression. Importantly, C6 subclones that expressed nestin at low levels in vitro were also found to give rise to tumors highly positive for the protein, suggesting that induction of nestin plays an important role in glioblastoma carcinogenesis. Derivation of nestin positive tumors from nestin negative human U87 glioblastoma cells in immunodeficient mice further confirmed that a switch to positive expression of nestin is fundamental to the course of glioblastoma development. Blocking the expression of nestin in glioblastoma tumors via intratumor injection of shRNA significantly slowed tumor growth and volume. These results demonstrated that nestin plays a crucial role in development of glioblastoma and may potentially be targeted for treatment of the disease.
Collapse
Affiliation(s)
- Wen Jing Lu
- Department of Cell Biology, Peking University Health Science Center, Beijing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Activation of STAT3 by Pituitary Adenylate Cyclase-Activating Polypeptide (PACAP) During PACAP-Promoted Neurite Outgrowth of PC12 Cells. J Mol Neurosci 2010; 42:349-58. [DOI: 10.1007/s12031-010-9376-3] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2009] [Accepted: 04/14/2010] [Indexed: 11/25/2022]
|
37
|
Li Y, Lu HM, Li G, Yan GM. Glycogen synthase kinase-3beta regulates astrocytic differentiation of U87-MG human glioblastoma cells. Acta Pharmacol Sin 2010; 31:355-60. [PMID: 20154711 DOI: 10.1038/aps.2010.10] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
AIM To evaluate the role of glycogen synthase kinase-3beta (GSK-3beta) in the induced differentiation of human glioblastoma cells. METHODS Cell proliferation was determined by bromodeoxyuridine (BrdU) incorporation assay. The protein level of p-GSK-3beta, GSK-3beta, glial fibrillary acidic protein (GFAP) and proliferating cell nuclear antigen (PCNA) were determined using Western blots. The overexpression of mutant GSK-3beta was analyzed by immunocytochemistry. RESULTS The biotoxin cholera toxin is capable of inducing differentiation of U87-MG human glioblastoma cells, which is characterized by morphological changes to astrocytic phenotype, increase in differentiation marker protein GFAP and decrease in proliferation. GSK-3beta activation is induced during this differentiation. Small interfering RNA against GSK-3beta suppresses the induced-differentiation in U87-MG cells. Conversely, overexpression of a constitutively active form of human GSK-3beta (pcDNA3-GSK-3beta-S9A) mutant leads to differentiation of U87-MG cells. CONCLUSION Our findings suggest that GSK-3beta plays an important role in astrocytic differentiation of human glioblastoma cells and may be a novel therapeutic target in the malignant tumor.
Collapse
|
38
|
Lu H, Li Y, Shu M, Tang J, Huang Y, Zhou Y, Liang Y, Yan G. Hypoxia-inducible factor-1α blocks differentiation of malignant gliomas. FEBS J 2009; 276:7291-304. [DOI: 10.1111/j.1742-4658.2009.07441.x] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/16/2023]
|
39
|
Murray PD, Kingsbury TJ, Krueger BK. Failure of Ca2+-activated, CREB-dependent transcription in astrocytes. Glia 2009; 57:828-34. [PMID: 19031446 DOI: 10.1002/glia.20809] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Astrocytes participate in signaling via Ca(2+) transients that spread from cell to cell across a multicellular syncytium. The effect, if any, of these Ca(2+) waves on the transcription of Ca(2+)/cAMP-regulatory element binding protein (CREB)-dependent genes is not known. We report here that, unlike neurons, increasing intracellular Ca(2+) in cultured mouse cortical astrocytes failed to activate CREB-dependent transcription, even though CREB was phosphorylated at serine 133. In contrast, both CREB phosphorylation and CREB-dependent transcription were robustly stimulated by increasing cAMP. The failure of Ca(2+)-activated transcription in astrocytes was correlated with the absence of CaMKIV, a Ca(2+)-dependent protein kinase required for Ca(2+)-stimulated gene transcription in neurons. The inability of Ca(2+) to signal via CaMKIV may insulate CREB-dependent gene transcription in astrocytes from activation by Ca(2+) waves.
Collapse
Affiliation(s)
- Peter D Murray
- Department of Physiology, University of Maryland School of Medicine, 655 W. Baltimore St., Baltimore, MD 21201, USA
| | | | | |
Collapse
|
40
|
Dziennis S, Alkayed NJ. Role of signal transducer and activator of transcription 3 in neuronal survival and regeneration. Rev Neurosci 2009; 19:341-61. [PMID: 19145989 DOI: 10.1515/revneuro.2008.19.4-5.341] [Citation(s) in RCA: 115] [Impact Index Per Article: 7.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Signal Transducers and Activators of Transcription (STATs) comprise a family of transcription factors that mediate a wide variety of biological functions in the central and peripheral nervous systems. Injury to neural tissue induces STAT activation, and STATs are increasingly recognized for their role in neuronal survival. In this review, we discuss the role of STAT3 during neural development and following ischemic and traumatic injury in brain, spinal cord and peripheral nerves. We focus on STAT3 because of the expanding body of literature that investigates protective and regenerative effects of growth factors, hormones and cytokines that use STAT3 to mediate their effect, in part through transcriptional upregulation of neuroprotective and neurotrophic genes. Defining the endogenous molecular mechanisms that lead to neuroprotection by STAT3 after injury might identify novel therapeutic targets against acute neural tissue damage as well as chronic neurodegenerative disorders.
Collapse
Affiliation(s)
- Suzan Dziennis
- Department of Anesthesiology & Peri-Operative Medicine, Oregon Health & Science University, Portland, OR 97239-3098, USA
| | | |
Collapse
|
41
|
Hai J, Lin Q, Lu Y. Phosphatidylinositol 3-kinase activity is required for the induction of differentiation in C6 glioma cells by panaxydol. J Clin Neurosci 2009; 16:444-8. [DOI: 10.1016/j.jocn.2008.05.014] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2008] [Revised: 05/13/2008] [Accepted: 05/18/2008] [Indexed: 11/16/2022]
|
42
|
Tatsumi K, Takebayashi H, Manabe T, Tanaka KF, Makinodan M, Yamauchi T, Makinodan E, Matsuyoshi H, Okuda H, Ikenaka K, Wanaka A. Genetic fate mapping of Olig2 progenitors in the injured adult cerebral cortex reveals preferential differentiation into astrocytes. J Neurosci Res 2008; 86:3494-502. [DOI: 10.1002/jnr.21862] [Citation(s) in RCA: 98] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
|
43
|
Hai J, Lin Q, Zhang H, Lu Y, Yi J. Cyclic AMP-dependent regulation of differentiation of rat C6 glioma cells by panaxydol. Neurol Res 2008; 31:274-9. [PMID: 19040798 DOI: 10.1179/174313209x380919] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/31/2022]
Abstract
OBJECTIVES Preliminary works have indicated that panaxydol possesses growth inhibition and induces differentiation in rat C6 glioma cells. However, the molecular mechanism underlying this differentiation remains unknown. We sought to investigate the role of cyclic adenosine monophosphate (cAMP) in cellular differentiation induced by panaxydol. METHODS C6 cells were treated with panaxydol and various specific inhibitors, and the inhibition of cell growth was assessed by 3-(4,5-dimethyl-2-thiazolyl)-2,5-diphenyl tetrazolium bromide assay and homotransplantation in nude mice. Astrocytic processes were quantified under a phase-contrasted microscope. Glial fibrillary acidic protein expression and cell migration were carried out by Western blot and scratch-wound test, respectively. In addition, the intracellular cAMP concentration was measured by immunoassay. RESULTS Panaxydol induces the elevation of intracellular cAMP concentration in C6 cells. The effects of growth inhibition in vitro and in vivo and induction of differentiation in C6 cells by panaxydol could be inhibited by the cAMP inhibitor, Rp-adenosine 3',5'-cyclic monophosphothioate, but not by protein kinase A or protein kinase C specific inhibitors. CONCLUSION These results suggest that the cAMP-dependent pathway may regulate cellular proliferation, migration and differentiation in C6 glioma cells by panaxydol.
Collapse
Affiliation(s)
- Jian Hai
- Department of Neurosurgery, Tongji Hospital, Tongji University, Shanghai, China
| | | | | | | | | |
Collapse
|
44
|
Tan JC, Li Y, Qu WY, Liu LY, Jiang L, Sun KL. Derivation of embryonic stem cell line from frozen human embryos and neural differentiation. Neuroreport 2008; 19:1451-5. [DOI: 10.1097/wnr.0b013e32830e4c35] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
45
|
|
46
|
Prostaglandin E1 inhibits IL-6-induced MCP-1 expression by interfering specifically in IL-6-dependent ERK1/2, but not STAT3, activation. Biochem J 2008; 412:65-72. [PMID: 18271757 DOI: 10.1042/bj20071572] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022]
Abstract
IL (interleukin)-6 exerts pro- as well as anti-inflammatory activities. Beside many other activities, IL-6 is the major inducer of acute phase proteins in the liver, acts as a differentiation factor for blood cells, as migration factor for T-cells and is a potent inducer of the chemokine MCP-1 (monocyte chemoattractant protein-1). Recent studies have focused on the negative regulation of IL-6 signal transduction through the IL-6-induced feedback inhibitors SOCS (suppressor of cytokine signalling) 1 and SOCS3 or the protein tyrosine phosphatases SHP-2 (Src homology 2 domain-containing protein tyrosine phosphatase 2) and TcPTP (T-cell protein tyrosine phosphatase). Studies on the cross-talk between pro-inflammatory mediators (IL-1, tumour necrosis factor, lipopolysaccharide) and IL-6 elucidated further regulatory mechanisms. Less is known about the regulation of IL-6 signal transduction by hormone/cytokine signalling through G-protein-coupled receptors. This is particularly surprising since many of these hormones (such as prostaglandins and chemokines) play an important role in inflammatory processes. In the present study, we have investigated the inhibitory activity of PGE(1) (prostaglandin E(1)) on IL-6-induced MCP-1 expression and have elucidated the underlying molecular mechanism. Surprisingly, PGE(1) does not affect IL-6-induced STAT (signal transducer and activator of transcription) 3 activation, but does affect ERK (extracellular-signal-regulated kinase) 1/2 activation which is crucial for IL-6-dependent expression of MCP-1. In summary, we have discovered a specific cross-talk between the adenylate cyclase cascade and the IL-6-induced MAPK (mitogen-activated protein kinase) cascade and have investigated its impact on IL-6-dependent gene expression.
Collapse
|
47
|
Tanabe K, Takai S, Matsushima-Nishiwaki R, Kato K, Dohi S, Kozawa O. Alpha2 adrenoreceptor agonist regulates protein kinase C-induced heat shock protein 27 phosphorylation in C6 glioma cells. J Neurochem 2008; 106:519-28. [PMID: 18384648 DOI: 10.1111/j.1471-4159.2008.05389.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Dexmedetomidine (Dexmd), a potent and highly specific alpha(2) adrenoreceptor agonist, is an efficient therapeutic agent for sedation. Dexmd has been recently reported to have a neuroprotective effect. Heat shock protein (HSP) 27, a low-molecular weight HSP has been shown to be expressed following cerebral ischemia in astrocytes but not in neurons. HSP27 expression is involved in ischemic tolerance of the brain. This study investigated the effect of Dexmd on HSP27 in rat C6 glioma cells. 12-O-tetradecanoylphorbol-13-actate (TPA), a direct activator of protein kinase C (PKC), stimulated the phosphorylation of HSP27 at Ser82, but not Ser15 in a time-dependent manner. Prostaglandin (PG) E(1) or PGE(2) which activates the adenylyl cyclase-cAMP system as well as forskolin and dibutyryl-cAMP, suppressed the TPA-induced phosphorylation of HSP27. Dexmd reversed the suppression of HSP27 phosphorylation by the adenylyl cyclase-cAMP system. Therefore, these results strongly suggest that Dexmd reverses the suppression of HSP27 phosphorylation by the adenylyl cyclase-cAMP system activation through the inhibition of its system in C6 cells. alpha(2) Adrenoreceptor agonists may therefore show a neuroprotective effect through the modification of HSP27 phosphorylation induced by PKC activation.
Collapse
Affiliation(s)
- Kumiko Tanabe
- Department of Anesthesiology and Pain Medicine, Gifu University Graduate School of Medicine, Gifu, Japan.
| | | | | | | | | | | |
Collapse
|
48
|
Makino N, Mise T, Sagara JI. Kinetics of hydrogen peroxide elimination by astrocytes and C6 glioma cells analysis based on a mathematical model. Biochim Biophys Acta Gen Subj 2008; 1780:927-36. [PMID: 18402782 DOI: 10.1016/j.bbagen.2008.03.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2008] [Revised: 02/19/2008] [Accepted: 03/13/2008] [Indexed: 10/22/2022]
Abstract
Oxidative stress is implicated in a variety of disorders including neurodegenerative diseases, and H(2)O(2) is important in the generation of reactive oxygen and oxidative stress. In this study, we have examined the rate of extracellular H(2)O(2) elimination and relevant enzyme activities in cultured astrocytes and C6 glioma cells and have analyzed the results based on a mathematical model. As compared with other types of cultured cells, astrocytes showed higher activity of glutathione peroxidase (GPx) but lower activities for GSH recycling. C6 cells showed relatively low GPx activity, and treatment of C6 cells with dibutyryl-cAMP, which induces astrocytic differentiation, increased catalase activity and H(2)O(2) permeation rate but exerted little effect on other enzyme activities. A mathematical model [N. Makino, K. Sasaki, N. Hashida, Y. Sakakura, A metabolic model describing the H(2)O(2) elimination by mammalian cells including H(2)O(2) permeation through cytoplasmic and peroxisomal membranes: comparison with experimental data, Biochim. Biophys. Acta 1673 (2004) 149-159.], which includes relevant enzymes and H(2)O(2) permeation through membranes, was found to be fitted well to the H(2)O(2) concentration dependences of removal reaction with the permeation rate constants as variable parameters. As compared with PC12 cells as a culture model for neuron, H(2)O(2) removal activity of astrocytes was considerably higher at physiological H(2)O(2) concentrations. The details of the mathematical model are presented in Appendix.
Collapse
Affiliation(s)
- Nobuo Makino
- Center for Humanity and Sciences, Ibaraki Prefectural University of Health Sciences, Ami 4669-2, Ami, Ibaraki 300-0394, Japan.
| | | | | |
Collapse
|
49
|
Mays CE, Kang HE, Kim Y, Shim SH, Bang JE, Woo HJ, Cho YH, Kim JB, Ryou C. CRBL cells: establishment, characterization and susceptibility to prion infection. Brain Res 2008; 1208:170-80. [PMID: 18395703 DOI: 10.1016/j.brainres.2008.02.103] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Revised: 02/07/2008] [Accepted: 02/10/2008] [Indexed: 11/16/2022]
Abstract
The cerebellum is involved in complex physiological functions including motor control, sensory perception, cognition, language, and emotion. Humans and animals with prion diseases are characterized clinically by ataxia, postural abnormalities and cognitive decline. Pathology in the cerebellum affected by prions includes spongiform degeneration, neuronal loss, and gliosis. To develop an in vitro model system for studying prion biology in cerebellar cells, we established and characterized an immortal cell line (CRBL) isolated from the cerebellum of mice lacking expression of a protein involved in cell cycle arrest. The characteristics of the cells include morphological heterogeneity, rapid proliferation, serum responsiveness during growth, and a change in the number of chromosomes. CRBL cells expressed both neuronal and glial cell markers as well as a considerable level of cellular prion protein, PrP(C). Upon in vitro infection, CRBL cells exhibited selective susceptibility to prions isolated from different sources. These cells chronically propagated prions from SMB cells. Strain-specific prion infection in CRBL cells was not due to instability of the cell line, allelic variance, or mutations in the PrP gene. Molecular properties of prions derived from SMB cells were maintained in the infected CRBL cells. Our results suggest that the specific interaction between a prion strain and hosts determined the selective susceptibility of CRBL cells, which reflects the conditions in vivo. In addition to the future studies revealing cellular and molecular mechanism involved in prion pathogenesis, CRBL cells will contribute to the studies dealing with prion strain properties and host susceptibilities.
Collapse
Affiliation(s)
- Charles E Mays
- Sanders Brown Center on Aging, Department of Microbiology, Immunology and Molecular Genetics, University of Kentucky College of Medicine, 800 Rose Street, HSRB-326, Lexington, KY 40536, USA
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Ito S, Nabetani T, Shinoda Y, Nagatsuka Y, Hirabayashi Y. Quantitative analysis of a novel glucosylated phospholipid by liquid chromatography-mass spectrometry. Anal Biochem 2008; 376:252-7. [PMID: 18342611 DOI: 10.1016/j.ab.2008.02.007] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2007] [Revised: 02/06/2008] [Accepted: 02/06/2008] [Indexed: 10/22/2022]
Abstract
Building upon the demonstrated presence of a new glyceroglycolipid, phosphatidylglucoside (PtdGlc), in rat embryonic brain tissues, we have developed a method to identify minute amounts of PtdGlc in cultured cells by using nano-flow high-performance liquid chromatography and negative-ion-mode electrospray linear-ion trap time-of-flight mass spectrometry (LC-MS). A normal-phase silica gel-based column enabled us to separate PtdGlc from other lipid classes. PtdGlc was identified from its tandem mass spectrometry spectrum and from its retention time in the column. Using an internal standard collection and LC-MS, we obtained the linearity of PtdGlc at a range of 6.3-800 fmol per injection. We applied this method to analyze quantitative changes in PtdGlc in C6 glioma cells after cellular differentiation into GFAP-positive glial cells. PtdGlc in C6 glioma cells consisted exclusively of C18:0/C20:0 fatty acyl chains. Differentiation induced by the addition of anti-PtdGlc antibody plus cAMP in culture medium significantly increased the glycolipid content.
Collapse
Affiliation(s)
- Shinya Ito
- Hitachi High-Technologies Corp., 1-24-14 Nishi-shinbashi, Minato-ku, Tokyo 105-8717, Japan
| | | | | | | | | |
Collapse
|