1
|
Kosenkov AM, Maiorov SA, Gaidin SG. Astrocytic NMDA Receptors. BIOCHEMISTRY. BIOKHIMIIA 2024; 89:1045-1060. [PMID: 38981700 DOI: 10.1134/s0006297924060063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/20/2023] [Revised: 04/24/2024] [Accepted: 04/25/2024] [Indexed: 07/11/2024]
Abstract
Astrocytic NMDA receptors (NMDARs) are heterotetramers, whose expression and properties are largely determined by their subunit composition. Astrocytic NMDARs are characterized by a low sensitivity to magnesium ions and low calcium conductivity. Their activation plays an important role in the regulation of various intracellular processes, such as gene expression and mitochondrial function. Astrocytic NMDARs are involved in calcium signaling in astrocytes and can act through the ionotropic and metabotropic pathways. Astrocytic NMDARs participate in the interactions of the neuroglia, thus affecting synaptic plasticity. They are also engaged in the astrocyte-vascular interactions and contribute to the regulation of vascular tone. Astrocytic NMDARs are involved in various pathologies, such as ischemia and hyperammonemia, and their blockade prevents negative changes in astrocytes during these diseases.
Collapse
Affiliation(s)
- Artem M Kosenkov
- Pushchino Scientific Center for Biological Research, Institute of Cell Biophysics of the Russian Academy of Sciences, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia.
| | - Sergei A Maiorov
- Pushchino Scientific Center for Biological Research, Institute of Cell Biophysics of the Russian Academy of Sciences, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| | - Sergei G Gaidin
- Pushchino Scientific Center for Biological Research, Institute of Cell Biophysics of the Russian Academy of Sciences, Russian Academy of Sciences, Pushchino, Moscow Region, 142290, Russia
| |
Collapse
|
2
|
Starinets A, Ponomarenko A, Tyrtyshnaia A, Manzhulo I. Synaptamide modulates glial and neurotransmitter activity in the spinal cord during neuropathic pain. J Chem Neuroanat 2023; 134:102361. [PMID: 37935251 DOI: 10.1016/j.jchemneu.2023.102361] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2023] [Revised: 11/03/2023] [Accepted: 11/03/2023] [Indexed: 11/09/2023]
Abstract
N-docosahexaenoylethanolamine, or synaptamide, is an endogenous metabolite of docosahexaenoic acid that is known for synaptogenic and neurogenic effects. In our previous studies we have shown that synaptamide attenuates neuropathic pain, facilitates remyelination, and reduces neuroinflammation after the chronic constriction injury (CCI) of the sciatic nerve in rats. In the current study, we show that daily synaptamide administration (4 mg/kg/day) within 14 days post-surgery: (1) decreases micro- and astroglia activity in the dorsal and ventral horns of the lumbar spinal cord; (2) modulates pro-inflammatory (IL1β, IL6) and anti-inflammatory (IL4, IL10) cytokine level in the serum and spinal cord; (3) leads to a rise in synaptamide and anandamide concentration in the spinal cord; (4) enhances IL10, CD206 and N-acylethanolamine-hydrolyzing acid amidase synthesis in macrophage cell culture following LPS-induced inflammation. Thus, the ability of synaptamide to modulate glial and cytokine activity indicates its potential for implementation in the treatment peripheral nerve injury.
Collapse
Affiliation(s)
- Anna Starinets
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Arina Ponomarenko
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Anna Tyrtyshnaia
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia
| | - Igor Manzhulo
- A.V. Zhirmunsky National Scientific Center of Marine Biology, Far Eastern Branch, Russian Academy of Sciences, Vladivostok 690041, Russia.
| |
Collapse
|
3
|
Shen Y, Lv F, Min S, Hao X, Yu J. Ketamine alleviating depressive-like behaviors is associated with regulation of nNOS-CAPON-Dexras1 complex in chronic unpredictable mild stress rats. Transl Neurosci 2022; 13:309-319. [PMID: 36212606 PMCID: PMC9508647 DOI: 10.1515/tnsci-2022-0245] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/29/2022] [Revised: 08/24/2022] [Accepted: 08/26/2022] [Indexed: 11/17/2022] Open
Abstract
BACKGROUND A growing number of studies have demonstrated that ketamine induces rapid and sustained antidepressant action. Neuronal nitric oxide synthase (nNOS) signaling has been explored for the treatment of neuropsychiatric disorders for decades. But the effect of ketamine on nNOS signaling is poorly understood. The aim of the present study was to investigate the effect of ketamine on nNOS signaling in a chronic unpredictable mild stress (CUMS) model of depression. METHODS Forty-eight rats were randomly divided into four groups: the control group of healthy rats (group C), the healthy rats treated with ketamine 10 mg/kg for 3 days (group CK), the rats model of stress-induced depression group (group D), and the depressed group treated with ketamine 10 mg/kg for 3 days (group DK). The sucrose preference test and open field test were used to assess behavioral changes. Immunohistochemistry, immunofluorescence, and real-time PCR analysis were carried out to measure the expression of nNOS, CAPON, and Dexras1 in the prefrontal cortex (PFC) of the CUMS rats. RESULTS Compared with healthy rats, the total distance traveled, the rearing counts, the sucrose preference percentage (SPP), and CAPON and Dexras1 expression in the PFC significantly decreased, while nNOS expression increased in CUMS rats. After treating with ketamine, the total distance traveled, the rearing counts, the SPP, and CAPON and Dexras1 expression significantly increased, while nNOS expression significantly decreased. CONCLUSION The results indicated that ketamine improved the depressive behavior of rats, which may be related to the reduced nNOS expression and enhanced CAPON and Dexras1 expression.
Collapse
Affiliation(s)
- Yiwei Shen
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Rd, Chongqing 400016, People’s Republic of China
| | - Feng Lv
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Rd, Chongqing 400016, People’s Republic of China
| | - Su Min
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Rd, Chongqing 400016, People’s Republic of China
| | - Xuechao Hao
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Rd, Chongqing 400016, People’s Republic of China
| | - Jian Yu
- Department of Anesthesiology, The First Affiliated Hospital of Chongqing Medical University, No. 1 Youyi Rd, Chongqing 400016, People’s Republic of China
| |
Collapse
|
4
|
Gu Y, Zhu D. nNOS-mediated protein-protein interactions: promising targets for treating neurological and neuropsychiatric disorders. J Biomed Res 2020; 35:1-10. [PMID: 33402546 PMCID: PMC7874267 DOI: 10.7555/jbr.34.20200108] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
Neurological and neuropsychiatric disorders are one of the leading causes of disability worldwide and affect the health of billions of people. Nitric oxide (NO), a free gas with multitudinous bioactivities, is mainly produced from the oxidation of L-arginine by neuronal nitric oxide synthase (nNOS) in the brain. Inhibiting nNOS benefits a variety of neurological and neuropsychiatric disorders, including stroke, depression and anxiety disorders, post-traumatic stress disorder, Parkinson's disease, Alzheimer's disease, chronic pain, and drug addiction. Due to critical roles of nNOS in learning and memory and synaptic plasticity, direct inhibition of nNOS may cause severe side effects. Importantly, interactions of several proteins, including post-synaptic density 95 (PSD-95), carboxy-terminal PDZ ligand of nNOS (CAPON) and serotonin transporter (SERT), with the PSD/Disc-large/ZO-1 homologous (PDZ) domain of nNOS have been demonstrated to influence the subcellular distribution and activity of the enzyme in the brain. Therefore, it will be a preferable means to interfere with nNOS-mediated protein-protein interactions (PPIs), which do not lead to undesirable effects. Herein, we summarize the current literatures on nNOS-mediated PPIs involved in neurological and neuropsychiatric disorders, and the discovery of drugs targeting the PPIs, which is expected to provide potential targets for developing novel drugs and new strategy for the treatment of neurological and neuropsychiatric disorders.
Collapse
Affiliation(s)
- Yuanyuan Gu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| | - Dongya Zhu
- Department of Pharmacology, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China.,Institution of Stem Cell and Neuroregeneration, School of Pharmacy, Nanjing Medical University, Nanjing, Jiangsu 211166, China
| |
Collapse
|
5
|
Gao S, Zhang T, Jin L, Liang D, Fan G, Song Y, Lucassen PJ, Yu R, Swaab DF. CAPON Is a Critical Protein in Synaptic Molecular Networks in the Prefrontal Cortex of Mood Disorder Patients and Contributes to Depression-Like Behavior in a Mouse Model. Cereb Cortex 2020; 29:3752-3765. [PMID: 30307500 DOI: 10.1093/cercor/bhy254] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/14/2018] [Revised: 08/16/2018] [Indexed: 12/16/2022] Open
Abstract
Aberrant regulation and activity of synaptic proteins may cause synaptic pathology in the prefrontal cortex (PFC) of mood disorder patients. Carboxy-terminal PDZ ligand of NOS1 (CAPON) is a critical scaffold protein linked to synaptic proteins like nitric oxide synthase 1, synapsins. We hypothesized that CAPON is altered together with its interacting synaptic proteins in the PFC in mood disorder patients and may contribute to depression-like behaviors in mice subjected to chronic unpredictable mild stress (CUMS). Here, we found that CAPON-immunoreactivity (ir) was significantly increased in the dorsolateral PFC (DLPFC) and anterior cingulate cortex in major depressive disorder (MDD), which was accompanied by an upregulation of spinophilin-ir and a downregulation of synapsin-ir. The increases in CAPON and spinophilin and the decrease in synapsin in the DLPFC of MDD patients were also seen in the PFC of CUMS mice. CAPON-ir positively correlated with spinophilin-ir (but not with synapsin-ir) in mood disorder patients. CAPON colocalized with spinophilin in the DLPFC of MDD patients and interacted with spinophilin in human brain. Viral-mediated CAPON downregulation in the medial PFC notably reversed the depression-like behaviors in the CUMS mice. These data suggest that CAPON may contribute to aspects of depressive behavior, possibly as an interacting protein for spinophilin in the PFC.
Collapse
Affiliation(s)
- Shangfeng Gao
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Tong Zhang
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Lei Jin
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Dong Liang
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Guangwei Fan
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Yunnong Song
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Paul J Lucassen
- Swammerdam Institute for Life Sciences, Center for Neuroscience, University of Amsterdam, Science Park 904, XH, Amsterdam, The Netherlands
| | - Rutong Yu
- Institute of Nervous System Diseases, Xuzhou Medical University, 84 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China.,Brain Hospital, The Affiliated Hospital of Xuzhou Medical University, 99 West Huai-Hai Road, Xuzhou, Jiangsu, P. R. China
| | - Dick F Swaab
- The Netherlands Institute for Neuroscience, Royal Netherlands Academy of Arts and Sciences, Meibergdreef 47, Amsterdam, The Netherlands
| |
Collapse
|
6
|
Skowrońska K, Obara-Michlewska M, Zielińska M, Albrecht J. NMDA Receptors in Astrocytes: In Search for Roles in Neurotransmission and Astrocytic Homeostasis. Int J Mol Sci 2019; 20:ijms20020309. [PMID: 30646531 PMCID: PMC6358855 DOI: 10.3390/ijms20020309] [Citation(s) in RCA: 73] [Impact Index Per Article: 12.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/28/2018] [Revised: 01/08/2019] [Accepted: 01/10/2019] [Indexed: 12/15/2022] Open
Abstract
Studies of the last two decades have demonstrated the presence in astrocytic cell membranes of N-methyl-d-aspartate (NMDA) receptors (NMDARs), albeit their apparently low abundance makes demonstration of their presence and function more difficult than of other glutamate (Glu) receptor classes residing in astrocytes. Activation of astrocytic NMDARs directly in brain slices and in acutely isolated or cultured astrocytes evokes intracellular calcium increase, by mutually unexclusive ionotropic and metabotropic mechanisms. However, other than one report on the contribution of astrocyte-located NMDARs to astrocyte-dependent modulation of presynaptic strength in the hippocampus, there is no sound evidence for the significant role of astrocytic NMDARs in astrocytic-neuronal interaction in neurotransmission, as yet. Durable exposure of astrocytic and neuronal co-cultures to NMDA has been reported to upregulate astrocytic synthesis of glutathione, and in this way to increase the antioxidative capacity of neurons. On the other hand, overexposure to NMDA decreases, by an as yet unknown mechanism, the ability of cultured astrocytes to express glutamine synthetase (GS), aquaporin-4 (AQP4), and the inward rectifying potassium channel Kir4.1, the three astroglia-specific proteins critical for homeostatic function of astrocytes. The beneficial or detrimental effects of astrocytic NMDAR stimulation revealed in the in vitro studies remain to be proven in the in vivo setting.
Collapse
Affiliation(s)
- Katarzyna Skowrońska
- Department of Neurotoxicology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106 Warsaw, Poland.
| | - Marta Obara-Michlewska
- Department of Neurotoxicology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106 Warsaw, Poland.
| | - Magdalena Zielińska
- Department of Neurotoxicology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106 Warsaw, Poland.
| | - Jan Albrecht
- Department of Neurotoxicology, Mossakowski Medical Research Centre, Polish Academy of Sciences, 5 Pawińskiego Str., 02-106 Warsaw, Poland.
| |
Collapse
|
7
|
Pierozan P, Biasibetti-Brendler H, Schmitz F, Ferreira F, Netto CA, Wyse ATS. Synergistic Toxicity of the Neurometabolites Quinolinic Acid and Homocysteine in Cortical Neurons and Astrocytes: Implications in Alzheimer's Disease. Neurotox Res 2017; 34:147-163. [PMID: 29124681 DOI: 10.1007/s12640-017-9834-6] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2017] [Revised: 09/22/2017] [Accepted: 10/18/2017] [Indexed: 12/19/2022]
Abstract
The brain of patients affected by Alzheimer's disease (AD) develops progressive neurodegeneration linked to the formation of proteins aggregates. However, their single actions cannot explain the extent of brain damage observed in this disorder, and the characterization of co-adjuvant involved in the early toxic processes evoked in AD is essential. In this line, quinolinic acid (QUIN) and homocysteine (Hcy) appear to be involved in the AD neuropathogenesis. Herein, we investigate the effects of QUIN and Hcy on early toxic events in cortical neurons and astrocytes. Exposure of primary cortical cultures to these neurometabolites for 24 h induced concentration-dependent neurotoxicity. In addition, QUIN (25 μM) and Hcy (30 μM) triggered ROS production, lipid peroxidation, diminished of Na+,K+-ATPase activity, and morphologic alterations, culminating in reduced neuronal viability by necrotic cell death. In astrocytes, QUIN (100 μM) and Hcy (30 μM) induced caspase-3-dependent apoptosis and morphologic alterations through oxidative status imbalance. To establish specific mechanisms, we preincubated cell cultures with different protective agents. The combined toxicity of QUIN and Hcy was attenuated by melatonin and Trolox in neurons and by NMDA antagonists and glutathione in astrocytes. Cellular death and morphologic alterations were prevented when co-culture was treated with metabolites, suggesting the activation of protector mechanisms dependent on soluble factors and astrocyte and neuron communication through gap junctions. These findings suggest that early damaging events involved in AD can be magnified by synergistic toxicity of the QUIN and Hcy. Therefore, this study opens new possibilities to elucidate the molecular mechanisms of neuron-astrocyte interactions and their role in neuroprotection against QUIN and Hcy.
Collapse
Affiliation(s)
- Paula Pierozan
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil.
| | - Helena Biasibetti-Brendler
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Felipe Schmitz
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Fernanda Ferreira
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
| | - Carlos Alexandre Netto
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Laboratório de Isquemia Cerebral e Psicobiologia dos Transtornos Mentais, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP, Porto Alegre, RS, 90035-003, Brazil
| | - Angela T S Wyse
- Laboratório de Neuroproteção e Doenças Metabólicas, Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos 2600 anexo, Porto Alegre, RS, 90035-003, Brazil
- Programa de Pós-Graduação em Ciências Biológicas: Bioquímica, Universidade Federal do Rio Grande do Sul, Porto Alegre, RS, Brazil
- Departamento de Bioquímica, Instituto de Ciências Básicas da Saúde, Universidade Federal do Rio Grande do Sul, Rua Ramiro Barcelos, 2600-Anexo, CEP, Porto Alegre, RS, 90035-003, Brazil
| |
Collapse
|
8
|
Abstract
Capon is a ligand protein of nitric oxide synthase 1. Recently, studies have shown that Capon is involved in the development of tumors. It is independent of the regulation of nitric oxide synthase 1 in this process. At the same time, studies have found that nitric oxide synthase 1 is expressed in multiple myeloma, but its role in the development and progression of myeloma remains unclear. In this study, we found that there was a different expression of Capon between the normal multiple myeloma cells and the adherent multiple myeloma cells. In the process of myeloma cell proliferation, the reduced expression of Capon reduces the arrest of the cell cycle in the G1 phase and promotes the proliferation of myeloma cells. Cell adhesion-mediated drug resistance is one of the most important factors, which affect the chemotherapy effect of multiple myeloma. If the expression of Capon is decreased, myeloma cells are adhered to fibronectin or bone marrow stromal cells (bone marrow mesenchymal stem cells). In addition, the sensitivity of the cell line to chemotherapeutic agents was reduced after silencing Capon in the myeloma cell line which was adhered to bone marrow mesenchymal stem cells. We also found that reduced expression of Capon resulted in the activation of the AKT signaling pathway. In conclusion, these results may be helpful in studying the role of Capon in multiple myeloma.
Collapse
Affiliation(s)
- Yaodong Shen
- 1 Department of Hematology, Affiliated Hospital of Nantong University, Nantong University, Nantong, P.R. China
| | - Haiyan Liu
- 1 Department of Hematology, Affiliated Hospital of Nantong University, Nantong University, Nantong, P.R. China
| | - Siyu Gu
- 1 Department of Hematology, Affiliated Hospital of Nantong University, Nantong University, Nantong, P.R. China
| | - Ziwei Wei
- 2 Nantong University, Nantong, P.R. China
| | - Hong Liu
- 1 Department of Hematology, Affiliated Hospital of Nantong University, Nantong University, Nantong, P.R. China
| |
Collapse
|
9
|
Low Expression of CAPON in Glioma Contributes to Cell Proliferation via the Akt Signaling Pathway. Int J Mol Sci 2016; 17:ijms17111859. [PMID: 27869735 PMCID: PMC5133859 DOI: 10.3390/ijms17111859] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/22/2016] [Revised: 10/27/2016] [Accepted: 11/01/2016] [Indexed: 12/25/2022] Open
Abstract
CAPON is an adapter protein for nitric oxide synthase 1 (NOS1). CAPON has two isoforms in the human brain: CAPON-L (long form of CAPON) and CAPON-S (short form of CAPON). Recent studies have indicated the involvement of CAPON in tumorigenesis beyond its classical role in NOS1 activity regulation. In this study, we found that the protein levels of CAPON-S, but not than CAPON-L, were significantly decreased in glioma tissues. Therefore, we established lentivirus-mediated stable cell lines with CAPON-S overexpression or down-regulation, and investigated the role of CAPON-S in the proliferation of glioma cells by using CCK8, EdU, and flow cytometry assays. Overexpression of CAPON-S reduced the cell variability and the percentage of EdU-positive cells, and arrested the cells in the G1 phase in glioma cells. Silencing of CAPON by short-hairpin RNA showed the opposite effects. Furthermore, an intracellular signaling array revealed that overexpression of CAPON-S resulted in a remarkable reduction in the phosphorylation of Akt and S6 ribosomal protein in glioma cells, which was further confirmed by Western blot. These findings suggest that CAPON may function as a tumor suppressor in human brain glioma and that the inactivation of the Akt signaling pathway caused by CAPON-S overexpression may provide insight into the underlying mechanism of CAPON in glioma cell proliferation.
Collapse
|
10
|
Pierozan P, Biasibetti H, Schmitz F, Ávila H, Fernandes CG, Pessoa-Pureur R, Wyse ATS. Neurotoxicity of Methylmercury in Isolated Astrocytes and Neurons: the Cytoskeleton as a Main Target. Mol Neurobiol 2016; 54:5752-5767. [DOI: 10.1007/s12035-016-0101-2] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2016] [Accepted: 09/05/2016] [Indexed: 01/16/2023]
|
11
|
Upregulation of PRDM5 Is Associated with Astrocyte Proliferation and Neuronal Apoptosis Caused by Lipopolysaccharide. J Mol Neurosci 2016; 59:146-57. [PMID: 27074744 DOI: 10.1007/s12031-016-0744-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/30/2016] [Accepted: 03/22/2016] [Indexed: 12/19/2022]
|
12
|
nNOS Translocates into the Nucleus and Interacts with Sox2 to Protect Neurons Against Early Excitotoxicity via Promotion of Shh Transcription. Mol Neurobiol 2015; 53:6444-6458. [PMID: 26607632 DOI: 10.1007/s12035-015-9545-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/20/2015] [Accepted: 11/16/2015] [Indexed: 10/25/2022]
Abstract
Cerebral ischemic stroke is a major public health problem leading to high mortality rates and disability in adults. The NMDA receptor (NMDAR)/neuronal nitric oxide synthase (nNOS)/NO-dependent excitotoxicity has been recognized to play an important role in cerebral ischemic stroke pathogenesis. Accumulating evidence suggests that the biological function of nNOS is associated with its ability to couple proteins and its subcellular localization. Previously, we and others determined that nNOS could translocate into the nucleus in cultured astrocytes, but the underlying mechanisms and biological significance remained unclear. In the present study, we identified a specific interaction between nNOS and Sox2 (SRY (sex determining region Y)-box 2), a member of the Sox family of transcription factors, both in vivo and in vitro. Our studies showed that nNOS is transported into the nucleus and interacted with Sox2 to form a nNOS-Sox2 complex in neurons at the early stage following glutamate stimulation. Mechanistically, via activating the transcription of Shh (Sonic hedgehog), the downstream target of Sox2, this nNOS-Sox2 complex exerted a neuroprotective function against glutamate-induced excitotoxicity. Utilizing the MCAO focal ischemia model on rats, we further verified that the 'nNOS-Sox2-Shh' axis was involved in the ischemic neuronal injury. Taken together, our studies revealed that the 'nNOS-Sox2-Shh' axis functions as a novel feedback compensatory mechanism to protect neurons against the early excitotoxicity and ischemic injury.
Collapse
|
13
|
Montes de Oca Balderas P, Aguilera P. A Metabotropic-Like Flux-Independent NMDA Receptor Regulates Ca2+ Exit from Endoplasmic Reticulum and Mitochondrial Membrane Potential in Cultured Astrocytes. PLoS One 2015; 10:e0126314. [PMID: 25954808 PMCID: PMC4425671 DOI: 10.1371/journal.pone.0126314] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2014] [Accepted: 03/31/2015] [Indexed: 01/22/2023] Open
Abstract
Astrocytes were long thought to be only structural cells in the CNS; however, their functional properties support their role in information processing and cognition. The ionotropic glutamate N-methyl D-aspartate (NMDA) receptor (NMDAR) is critical for CNS functions, but its expression and function in astrocytes is still a matter of research and debate. Here, we report immunofluorescence (IF) labeling in rat cultured cortical astrocytes (rCCA) of all NMDAR subunits, with phenotypes suggesting their intracellular transport, and their mRNA were detected by qRT-PCR. IF and Western Blot revealed GluN1 full-length synthesis, subunit critical for NMDAR assembly and transport, and its plasma membrane localization. Functionally, we found an iCa2+ rise after NMDA treatment in Fluo-4-AM labeled rCCA, an effect blocked by the NMDAR competitive inhibitors D(-)-2-amino-5-phosphonopentanoic acid (APV) and Kynurenic acid (KYNA) and dependent upon GluN1 expression as evidenced by siRNA knock down. Surprisingly, the iCa2+ rise was not blocked by MK-801, an NMDAR channel blocker, or by extracellular Ca2+ depletion, indicating flux-independent NMDAR function. In contrast, the IP3 receptor (IP3R) inhibitor XestosponginC did block this response, whereas a Ryanodine Receptor inhibitor did so only partially. Furthermore, tyrosine kinase inhibition with genistein enhanced the NMDA elicited iCa2+ rise to levels comparable to those reached by the gliotransmitter ATP, but with different population dynamics. Finally, NMDA depleted the rCCA mitochondrial membrane potential (mΔψ) measured with JC-1. Our results demonstrate that rCCA express NMDAR subunits which assemble into functional receptors that mediate a metabotropic-like, non-canonical, flux-independent iCa2+ increase.
Collapse
Affiliation(s)
- Pavel Montes de Oca Balderas
- Unidad de Neurobiología Dinámica, Departamento de Neuroquímica, Instituto Nacional de Neurología y Neurocirugía, México City, México
- * E-mail:
| | - Penélope Aguilera
- Laboratorio de Patología Vascular Cerebral, Instituto Nacional de Neurología y Neurocirugía, México City, México
| |
Collapse
|
14
|
Cao M, Tan X, Jin W, Zheng H, Xu W, Rui Y, Li L, Cao J, Wu X, Cui G, Ke K, Gao Y. Upregulation of Ras homolog enriched in the brain (Rheb) in lipopolysaccharide-induced neuroinflammation. Neurochem Int 2013; 62:406-17. [PMID: 23391520 DOI: 10.1016/j.neuint.2013.01.025] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2012] [Revised: 12/16/2012] [Accepted: 01/26/2013] [Indexed: 12/16/2022]
Abstract
Ras homolog enriched in the brain (Rheb) is a homolog of Ras GTPase that regulates cell growth, proliferation, and cell cycle via mammalian target of rapamycin (mTOR). Recently, it has been confirmed that Rheb activation not only promotes cellular proliferation and differentiation but also enhances cellular apoptosis in response to diverse toxic stimuli. However, the function of Rheb in the central nervous system (CNS) is still with limited understanding. To elaborate whether Rheb was involved in CNS injury, we performed a neuroinflammatory model by lipopolysaccharide (LPS) lateral ventral injection in adult rats. Upregulation of Rheb was observed in the brain cortex by performing western blotting and immunohistochemistry. Double immunofluorescent staining demonstrated that Rheb was mainly in active astrocytes and neurons. PCNA and active caspase-3 were upregulated, and co-labeling with Rheb, which indicated that Rheb might be relevant to astrocytic proliferation and neuronal apoptosis following the inflammatory response by LPS-induced. Furthermore, we also found that the expression profiles of cyclinD1 and CDK4 were parallel with that of Rheb in a time-space dependent manner. Finally, knocking down Rheb by siRNA and treatment with rapamycin or lovastatin showed that not only astrocytic proliferation decreased but also neuronal protection. Based on our data, we suggested that Rheb might play an important role in physiological and pathological functions following neuroinflammation caused by LPS, which might provide a potential target to the treatment of neuroinflammation.
Collapse
Affiliation(s)
- Maohong Cao
- Department of Neurology, Affiliated Hospital of Nantong University, Nantong, Jiangsu Province 226001, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|