1
|
Deng B, He X, Wang Z, Kang J, Zhang G, Li L, Kang X. HSP70 protects PC12 cells against TBHP-induced apoptosis and oxidative stress by activating the Nrf2/HO-1 signaling pathway. In Vitro Cell Dev Biol Anim 2024:10.1007/s11626-024-00924-0. [PMID: 38807023 DOI: 10.1007/s11626-024-00924-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2024] [Accepted: 05/13/2024] [Indexed: 05/30/2024]
Abstract
HSP70 exhibits neuroprotective, antioxidant, and anti-apoptotic properties, which are crucial in preventing spinal cord injury (SCI) induced by oxidative stress and apoptosis. In this study, we assessed the potential protective effects and underlying mechanisms of HSP70 on tert-butyl hydroperoxide (TBHP)-damaged PC12 cells in an in vitro model of SCI. To establish the model, PC12 cells were subjected to oxidative damage induced by TBHP, followed by overexpression of HSP70. Cell viability was assessed using the CCK8 kit, intracellular reactive oxygen species level was evaluated using a commercial kit, cell apoptosis was detected using the Annexin V-APC/7-ADD Apoptosis Detection Kit, and the oxidative stress level was determined using SOD and MDA assay kits. Western blot analysis was used to detect the expression levels of Bax, cleaved caspase-3, and Bcl-2 proteins. Furthermore, immunofluorescence staining and Western bolt were used to detect the expression levels of proteins associated with the Nrf2/HO-1 signaling pathway. We found that HSP70 overexpression reduced apoptosis and oxidative stress in TBHP-induced PC12 cells. Furthermore, it activated the Nrf2/HO-1 signaling pathway. In addition, the Nrf2 inhibitor ML385 attenuated the protective effects of HSP70 on TBHP-induced PC12 cells. In conclusion, HSP70 can partially alleviate TBHP-induced apoptosis and oxidative stress in PC12 cells by promoting the Nrf2/HO-1 signaling pathway.
Collapse
Affiliation(s)
- Bo Deng
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Xuegang He
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Zhaoheng Wang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Jihe Kang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Guangzhi Zhang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Lei Li
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730000, China
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China
| | - Xuewen Kang
- Department of Orthopedics, Lanzhou University Second Hospital, Lanzhou, 730000, China.
- Key Laboratory of Orthopedics Disease of Gansu Province, Lanzhou University Second Hospital, Lanzhou, 730030, China.
| |
Collapse
|
2
|
Li Z, Liu H, Han W, Zhu S, Liu C. NMN Alleviates NP-Induced Learning and Memory Impairment Through SIRT1 Pathway in PC-12 Cell. Mol Neurobiol 2023; 60:2871-2883. [PMID: 36745337 DOI: 10.1007/s12035-023-03251-9] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2022] [Accepted: 01/28/2023] [Indexed: 02/07/2023]
Abstract
Nonylphenol (NP) is widely used in the chemical industry; it accumulates in organisms through environmental contamination and causes learning memory impairment. Nicotinamide mononucleotide (NMN) has been found to have a positive effect on the treatment of central nervous-related diseases. This study aimed to investigate the protective effect of NMN on NP-induced learning memory-related impairment in vitro and to further identify the underlying mechanisms. The results showed that NP induced oxidative stress and impaired the cholinergic system, 5-HT system in PC-12 cells. NMN alleviated NP-induced learning and memory impairment at the molecular level through alleviating oxidative stress and protective effects on the 5-HT system and cholinergic system. The 50 μM NP group significantly reduced the NAD+ content, and the relative expression of SIRT1, PGC-1α, Nrf2, MAOA, BDNF, and p-TrkB were significantly downregulated. Co-treatment of NMN with NP significantly reduced oxidative stress, improved the homeostasis of 5-HT and cholinergic system, enhanced the intracellular NAD+ content, and significantly upregulated the expression of SIRT1 pathway proteins. SIRT1 inhibitors reduced the expression of SIRT1 pathway-related proteins, which implied the impairment of learning and memory by NP and the protective effect of NMN might be achieved through the SIRT1-mediated PGC-1α/MAOA/BDNF signaling pathway. Overall, this study not only help us to understand the toxic mechanism of NP on learning memory impairment in vitro, but also have important reference significance to further explore the health care value of NMN and promote the development of related functional foods.
Collapse
Affiliation(s)
- Zhongyi Li
- College of Food Science, South China Agricultural University, 483#, Wu-Shan Ave., Tian-He District, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Food Quality and Safety, Guangzhou, 510642, China
| | - Huan Liu
- College of Food Science, South China Agricultural University, 483#, Wu-Shan Ave., Tian-He District, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Food Quality and Safety, Guangzhou, 510642, China
| | - Wenna Han
- College of Food Science, South China Agricultural University, 483#, Wu-Shan Ave., Tian-He District, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Food Quality and Safety, Guangzhou, 510642, China
| | - Siyu Zhu
- College of Food Science, South China Agricultural University, 483#, Wu-Shan Ave., Tian-He District, Guangzhou, 510642, China
- Guangdong Provincial Key Laboratory of Food Quality and Safety, Guangzhou, 510642, China
| | - Chunhong Liu
- College of Food Science, South China Agricultural University, 483#, Wu-Shan Ave., Tian-He District, Guangzhou, 510642, China.
- Guangdong Provincial Key Laboratory of Food Quality and Safety, Guangzhou, 510642, China.
| |
Collapse
|
3
|
Delage C, Breard-Mellin L, Thérésine C, Simioneck S, Lefranc B, Leprince J, Bénard M, Vaudry D. The Heterogeneity of Response of PC12 Cells from Different Laboratories to Nerve Growth Factor and Pituitary Adenylate Cyclase-Activating Polypeptide Questions the Reproducibility of Studies Carried Out with Tumor Cell Lines. Neuroendocrinology 2023; 113:216-230. [PMID: 34348336 DOI: 10.1159/000518337] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/24/2020] [Accepted: 07/05/2021] [Indexed: 11/19/2022]
Abstract
BACKGROUND PC12 pheochromocytoma tumor cell lines are widely used to decipher the intracellular signaling mechanisms mediating the effects of some growth factors. Nevertheless, the disparity in appearance of some PC12 cell lines used in the different publications questions our ability to compare the results obtained by the numerous laboratories which use them. This led us to analyze the phenotypic aspect and transcriptomic expression of 5 PC12 cell lines from different origins under control conditions and after treatment with nerve growth factor (NGF) or pituitary adenylate cyclase-activating polypeptide (PACAP). METHODS Characterization of the 5 PC12 cell lines was conducted using imaging techniques and high-throughput real-time PCR combined with bioinformatics analysis. RESULTS The results show that the 5 cell lines are very variable in terms of shape, proliferation rate, motility, adhesion to the substrate, and gene expression. This high heterogeneity of the cell lines is also found when looking at their response to NGF or PACAP on gene expression or differentiation, with even in some cases opposite effects, as, for example, on cell proliferation. Actually, only 2 of the cell lines tested exhibited some phenotypic similarities with each other, even though the transcriptomic analyses show that they are far from identical. DISCUSSION/CONCLUSION As this issue of cell heterogenicity is not restricted to PC12 cells, the present results highlight the need to facilitate the supply of cell lines at low cost, the necessity to standardize practices regarding the use of cell lines, and the requirement to define precise markers of established cell lines which should be monitored in every publication. Regarding this latter point, the present data show that transcriptomic analysis by real-time PCR using a panel of genes of interest is easy to implement and provides a reliable method to control the possible drift of the cells over time in culture. Transcriptomic phenotyping combined with bioinformatics analysis can also be a useful approach to predict the response of the cells to treatments in terms of cell signaling activation, which can help to choose among several cell lines the most appropriate one for the investigation of a particular mechanism. Taken together, the results from this study highlight the need to use well-characterized cell lines with standardized protocols to generate reproducible results from 1 laboratory to the other.
Collapse
Affiliation(s)
- Colombe Delage
- UNIROUEN, Inserm U1239, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal death and Cell plasticity team, Normandie University, Rouen, France
| | - Lou Breard-Mellin
- UNIROUEN, Inserm U1239, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal death and Cell plasticity team, Normandie University, Rouen, France
| | - Caroline Thérésine
- UNIROUEN, Inserm, Regional Cell Imaging Platform of Normandy (PRIMACEN), Institute for Research and Innovation in Biomedicine (IRIB), Normandie University, Rouen, France
| | - Séphora Simioneck
- UNIROUEN, Inserm U1239, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal death and Cell plasticity team, Normandie University, Rouen, France
| | - Benjamin Lefranc
- UNIROUEN, Inserm U1239, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal death and Cell plasticity team, Normandie University, Rouen, France
- UNIROUEN, Inserm, Regional Cell Imaging Platform of Normandy (PRIMACEN), Institute for Research and Innovation in Biomedicine (IRIB), Normandie University, Rouen, France
| | - Jérôme Leprince
- UNIROUEN, Inserm U1239, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal death and Cell plasticity team, Normandie University, Rouen, France
- UNIROUEN, Inserm, Regional Cell Imaging Platform of Normandy (PRIMACEN), Institute for Research and Innovation in Biomedicine (IRIB), Normandie University, Rouen, France
| | - Magalie Bénard
- UNIROUEN, Inserm, Regional Cell Imaging Platform of Normandy (PRIMACEN), Institute for Research and Innovation in Biomedicine (IRIB), Normandie University, Rouen, France
| | - David Vaudry
- UNIROUEN, Inserm U1239, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal death and Cell plasticity team, Normandie University, Rouen, France
- UNIROUEN, Inserm, Regional Cell Imaging Platform of Normandy (PRIMACEN), Institute for Research and Innovation in Biomedicine (IRIB), Normandie University, Rouen, France
| |
Collapse
|
4
|
Jin L, Wang Q, Yang M, Zhang J, Liang H, Tan H, Liang Z, Ma X, Liu J, Li H, Cai X, Cui W, Zhao L. Indirubin-3′-monoxime-loaded PLGA-PEG nanoparticles for potential Alzheimer's disease treatment. MEDICINE IN NOVEL TECHNOLOGY AND DEVICES 2022. [DOI: 10.1016/j.medntd.2022.100150] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
|
5
|
Mattei AM, Smailys JD, Hepworth EMW, Hinton SD. The Roles of Pseudophosphatases in Disease. Int J Mol Sci 2021; 22:ijms22136924. [PMID: 34203203 PMCID: PMC8269279 DOI: 10.3390/ijms22136924] [Citation(s) in RCA: 12] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2021] [Revised: 06/10/2021] [Accepted: 06/24/2021] [Indexed: 01/07/2023] Open
Abstract
The pseudophosphatases, atypical members of the protein tyrosine phosphatase family, have emerged as bona fide signaling regulators within the past two decades. Their roles as regulators have led to a renaissance of the pseudophosphatase and pseudoenyme fields, catapulting interest from a mere curiosity to intriguing and relevant proteins to investigate. Pseudophosphatases make up approximately fourteen percent of the phosphatase family, and are conserved throughout evolution. Pseudophosphatases, along with pseudokinases, are important players in physiology and pathophysiology. These atypical members of the protein tyrosine phosphatase and protein tyrosine kinase superfamily, respectively, are rendered catalytically inactive through mutations within their catalytic active signature motif and/or other important domains required for catalysis. This new interest in the pursuit of the relevant functions of these proteins has resulted in an elucidation of their roles in signaling cascades and diseases. There is a rapid accumulation of knowledge of diseases linked to their dysregulation, such as neuropathies and various cancers. This review analyzes the involvement of pseudophosphatases in diseases, highlighting the function of various role(s) of pseudophosphatases involvement in pathologies, and thus providing a platform to strongly consider them as key therapeutic drug targets.
Collapse
|
6
|
Mohamedi Y, Fontanil T, Cobo T, Cal S, Obaya AJ. New Insights into ADAMTS Metalloproteases in the Central Nervous System. Biomolecules 2020; 10:biom10030403. [PMID: 32150898 PMCID: PMC7175268 DOI: 10.3390/biom10030403] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2020] [Revised: 02/24/2020] [Accepted: 03/02/2020] [Indexed: 12/13/2022] Open
Abstract
Components of the extracellular matrix (ECM) are key players in regulating cellular functions throughout the whole organism. In fact, ECM components not only participate in tissue organization but also contribute to processes such as cellular maintenance, proliferation, and migration, as well as to support for various signaling pathways. In the central nervous system (CNS), proteoglycans of the lectican family, such as versican, aggrecan, brevican, and neurocan, are important constituents of the ECM. In recent years, members of this family have been found to be involved in the maintenance of CNS homeostasis and to participate directly in processes such as the organization of perineural nets, the regulation of brain plasticity, CNS development, brain injury repair, axonal guidance, and even the altering of synaptic responses. ADAMTSs are a family of “A disintegrin and metalloproteinase with thrombospondin motifs” proteins that have been found to be involved in a multitude of processes through the degradation of lecticans and other proteoglycans. Recently, alterations in ADAMTS expression and activity have been found to be involved in neuronal disorders such as stroke, neurodegeneration, schizophrenia, and even Alzheimer’s disease, which in turn may suggest their potential use as therapeutic targets. Herein, we summarize the different roles of ADAMTSs in regulating CNS events through interactions and the degradation of ECM components (more specifically, the lectican family of proteoglycans).
Collapse
Affiliation(s)
- Yamina Mohamedi
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain; (Y.M.); (T.F.); (S.C.)
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
| | - Tania Fontanil
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain; (Y.M.); (T.F.); (S.C.)
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- Departamento de Investigación, Instituto Ordóñez, 33012 Oviedo, Asturias, Spain
| | - Teresa Cobo
- Departamento de Cirugía y Especialidades Médico-Quirúrgicas, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain;
- Instituto Asturiano de Odontología, 33006 Oviedo, Asturias, Spain
| | - Santiago Cal
- Departamento de Bioquímica y Biología Molecular, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain; (Y.M.); (T.F.); (S.C.)
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
| | - Alvaro J. Obaya
- Departamento de Biología Funcional, Área de Fisiología, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- Instituto Universitario de Oncología, IUOPA, Universidad de Oviedo, 33006 Oviedo, Asturias, Spain
- Correspondence:
| |
Collapse
|
7
|
Corbière A, Vaudry H, Chan P, Walet-Balieu ML, Lecroq T, Lefebvre A, Pineau C, Vaudry D. Strategies for the Identification of Bioactive Neuropeptides in Vertebrates. Front Neurosci 2019; 13:948. [PMID: 31619945 PMCID: PMC6759750 DOI: 10.3389/fnins.2019.00948] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2019] [Accepted: 08/22/2019] [Indexed: 11/13/2022] Open
Abstract
Neuropeptides exert essential functions in animal physiology by controlling e.g., reproduction, development, growth, energy homeostasis, cardiovascular activity and stress response. Thus, identification of neuropeptides has been a very active field of research over the last decades. This review article presents the various methods used to discover novel bioactive peptides in vertebrates. Initially identified on the basis of their biological activity, some neuropeptides have also been discovered for their ability to bind/activate a specific receptor or based on their biochemical characteristics such as C-terminal amidation which concerns half of the known neuropeptides. More recently, sequencing of the genome of many representative species has facilitated peptidomic approaches using mass spectrometry and in silico screening of genomic libraries. Through these different approaches, more than a hundred of bioactive neuropeptides have already been identified in vertebrates. Nevertheless, researchers continue to find new neuropeptides or to identify novel functions of neuropeptides that had not been detected previously, as it was recently the case for nociceptin.
Collapse
Affiliation(s)
- Auriane Corbière
- Normandie Univ, UNIROUEN, Inserm, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, Rouen, France
| | - Hubert Vaudry
- Normandie Univ, UNIROUEN, Inserm, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, Rouen, France.,Normandie Univ, UNIROUEN, Regional Cell Imaging Platform of Normandy (PRIMACEN), Rouen, France
| | - Philippe Chan
- Normandie Univ, UNIROUEN, Rouen Proteomic Platform (PISSARO), Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Marie-Laure Walet-Balieu
- Normandie Univ, UNIROUEN, Rouen Proteomic Platform (PISSARO), Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| | - Thierry Lecroq
- Normandie Univ, UNIROUEN, LITIS EA 4108, Information Processing in Biology & Health, Rouen, France
| | - Arnaud Lefebvre
- Normandie Univ, UNIROUEN, LITIS EA 4108, Information Processing in Biology & Health, Rouen, France
| | | | - David Vaudry
- Normandie Univ, UNIROUEN, Inserm, Laboratory of Neuronal and Neuroendocrine Communication and Differentiation, Neuropeptides, Neuronal Death and Cell Plasticity Team, Rouen, France.,Normandie Univ, UNIROUEN, Regional Cell Imaging Platform of Normandy (PRIMACEN), Rouen, France.,Normandie Univ, UNIROUEN, Rouen Proteomic Platform (PISSARO), Institute for Research and Innovation in Biomedicine (IRIB), Rouen, France
| |
Collapse
|
8
|
Chen W, Deng W, Goldys EM. Light-Triggerable Liposomes for Enhanced Endolysosomal Escape and Gene Silencing in PC12 Cells. MOLECULAR THERAPY. NUCLEIC ACIDS 2017. [PMID: 28624212 PMCID: PMC5423320 DOI: 10.1016/j.omtn.2017.04.015] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
Abstract
Liposomes are an effective gene and/or drug delivery system, widely used in biomedical applications including gene therapy and chemotherapy. Here, we designed a photo-responsive liposome (lipVP) loaded with a photosensitizer verteporfin (VP). This photosensitizer is clinically approved for photodynamic therapy (PDT). LipVP was employed as a DNA carrier for pituitary adenylyl cyclase-activating polypeptide (PACAP) receptor 1 (PAC1R) gene knockdown in PC12 cells. This has been done by incorporating PAC1R antisense oligonucleotides inside the lipVP cavity. Cells that have taken up the lipVP were exposed to light from a UV light source. As a result of this exposure, reactive oxygen species (ROS) were generated from VP, destabilizing the endolysosomal membranes and enhancing the liposomal release of antisense DNA into the cytoplasm. Endolysosomal escape of DNA was documented at different time points based on quantitative analysis of colocalization between fluorescently labeled DNA and endosomes and lysosomes. The released antisense oligonucleotides were found to silence PAC1R mRNA. The efficiency of this photo-induced gene silencing was demonstrated by a 74% ± 5% decrease in PAC1R fluorescence intensity. Following the light-induced DNA transfer into cells, cell differentiation with exposure to two kinds of PACAP peptides was observed to determine the cell phenotypic change after PAC1R gene knockdown.
Collapse
Affiliation(s)
- Wenjie Chen
- ARC Centre of Excellence for Nanoscale BioPhotonics, Department of Physics and Astronomy, Macquarie University, Sydney, NSW 2109, Australia
| | - Wei Deng
- ARC Centre of Excellence for Nanoscale BioPhotonics, Department of Physics and Astronomy, Macquarie University, Sydney, NSW 2109, Australia.
| | - Ewa M Goldys
- ARC Centre of Excellence for Nanoscale BioPhotonics, Department of Physics and Astronomy, Macquarie University, Sydney, NSW 2109, Australia.
| |
Collapse
|
9
|
Liu C, Sun Y, Song Y, Saito T, Kurasaki M. Nonylphenol diethoxylate inhibits apoptosis induced in PC12 cells. ENVIRONMENTAL TOXICOLOGY 2016; 31:1389-1398. [PMID: 25847295 DOI: 10.1002/tox.22144] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 08/29/2014] [Revised: 03/13/2015] [Accepted: 03/18/2015] [Indexed: 06/04/2023]
Abstract
Nonylphenol and short-chain nonylphenol ethoxylates such as NP2 EO are present in aquatic environment as wastewater contaminants, and their toxic effects on aquatic species have been reported. Apoptosis has been shown to be induced by serum deprivation or copper treatment. To understand the toxicity of nonylphenol diethoxylate, we investigated the effects of NP2 EO on apoptosis induced by serum deprivation and copper by using PC12 cell system. Nonylphenol diethoxylate itself showed no toxicity and recovered cell viability from apoptosis. In addition, nonylphenol diethoxylate decreased DNA fragmentation caused by apoptosis in PC12 cells. This phenomenon was confirmed after treating apoptotic PC12 cells with nonylphenol diethoxylate, whereas the cytochrome c release into the cytosol decreased as compared to that in apoptotic cells not treated with nonylphenol diethoxylates. Furthermore, Bax contents in apoptotic cells were reduced after exposure to nonylphenol diethoxylate. Thus, nonylphenol diethoxylate has the opposite effect on apoptosis in PC12 cells compared to nonylphenol, which enhances apoptosis induced by serum deprivation. The difference in structure of the two compounds is hypothesized to be responsible for this phenomenon. These results indicated that nonylphenol diethoxylate has capability to affect cell differentiation and development and has potentially harmful effect on organisms because of its unexpected impact on apoptosis. © 2015 Wiley Periodicals, Inc. Environ Toxicol 31: 1389-1398, 2016.
Collapse
Affiliation(s)
- Chuang Liu
- Graduate School of Environmental Science, Hokkaido University, Sapporo, 060-0810, Japan
| | - Yongkun Sun
- Department of Basic Medicine, Xinxiang Medical University, Xinxiang, Henan, 453-003, China
- Faculty of Environmental Earth Science, Hokkaido University, Sapporo, 060-0810, Japan
| | - Yutong Song
- Graduate School of Environmental Science, Hokkaido University, Sapporo, 060-0810, Japan
| | - Takeshi Saito
- Faculty of Health Sciences, Hokkaido University, Sapporo, 060-0812, Japan
| | - Masaaki Kurasaki
- Graduate School of Environmental Science, Hokkaido University, Sapporo, 060-0810, Japan.
- Faculty of Environmental Earth Science, Hokkaido University, Sapporo, 060-0810, Japan.
| |
Collapse
|
10
|
Seaborn T, Ravni A, Au R, Chow BKC, Fournier A, Wurtz O, Vaudry H, Eiden LE, Vaudry D. Induction of serpinb1a by PACAP or NGF is required for PC12 cells survival after serum withdrawal. J Neurochem 2014; 131:21-32. [PMID: 24899316 DOI: 10.1111/jnc.12780] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 04/30/2014] [Accepted: 05/13/2014] [Indexed: 01/11/2023]
Abstract
PC12 cells are used to study the signaling mechanisms underlying the neurotrophic and neuroprotective activities of pituitary adenylate cyclase-activating polypeptide (PACAP) and nerve growth factor (NGF). Previous microarray experiments indicated that serpinb1a was the most induced gene after 6 h of treatment with PACAP or NGF. This study confirmed that serpinb1a is strongly activated by PACAP and NGF in a time-dependent manner with a maximum induction (~ 50-fold over control) observed after 6 h of treatment. Co-incubation with PACAP and NGF resulted in a synergistic up-regulation of serpinb1a expression (200-fold over control), suggesting that PACAP and NGF act through complementary mechanisms. Consistently, PACAP-induced serpinb1a expression was not blocked by TrkA receptor inhibition. Nevertheless, the stimulation of serpinb1a expression by PACAP and NGF was significantly reduced in the presence of extracellular signal-regulated kinase, calcineurin, protein kinase A, p38, and PI3K inhibitors, indicating that the two trophic factors share some common pathways in the regulation of serpinb1a. Finally, functional investigations conducted with siRNA revealed that serpinb1a is not involved in the effects of PACAP and NGF on PC12 cell neuritogenesis, proliferation or body cell volume but mediates their ability to block caspases 3/7 activity and to promote PC12 cell survival.
Collapse
Affiliation(s)
- Tommy Seaborn
- Neurotrophic Factor and Neuronal Differentiation Team, Inserm U982, DC2N, Mont-Saint-Aignan, France; International Associated Laboratory Samuel de Champlain, Mont-Saint-Aignan, France; Department of Pediatrics, Hôpital St-François d'Assise, Centre de Recherche du Centre Hospitalier Universitaire de Québec (CRCHUQ), Laval University, Québec, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Krock E, Rosenzweig DH, Chabot-Doré AJ, Jarzem P, Weber MH, Ouellet JA, Stone LS, Haglund L. Painful, degenerating intervertebral discs up-regulate neurite sprouting and CGRP through nociceptive factors. J Cell Mol Med 2014; 18:1213-25. [PMID: 24650225 PMCID: PMC4508160 DOI: 10.1111/jcmm.12268] [Citation(s) in RCA: 114] [Impact Index Per Article: 11.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2013] [Accepted: 01/29/2014] [Indexed: 01/07/2023] Open
Abstract
Intervertebral disc degeneration (IVD) can result in chronic low back pain, a common cause of morbidity and disability. Inflammation has been associated with IVD degeneration, however the relationship between inflammatory factors and chronic low back pain remains unclear. Furthermore, increased levels of nerve growth factor (NGF) and brain derived neurotrophic factor (BDNF) are both associated with inflammation and chronic low back pain, but whether degenerating discs release sufficient concentrations of factors that induce nociceptor plasticity remains unclear. Degenerating IVDs from low back pain patients and healthy, painless IVDs from human organ donors were cultured ex vivo. Inflammatory and nociceptive factors released by IVDs into culture media were quantified by enzyme-linked immunosorbent assays and protein arrays. The ability of factors released to induce neurite growth and nociceptive neuropeptide production was investigated. Degenerating discs release increased levels of tumour necrosis factor-α, interleukin-1β, NGF and BDNF. Factors released by degenerating IVDs increased neurite growth and calcitonin gene-related peptide expression, both of which were blocked by anti-NGF treatment. Furthermore, protein arrays found increased levels of 20 inflammatory factors, many of which have nociceptive effects. Our results demonstrate that degenerating and painful human IVDs release increased levels of NGF, inflammatory and nociceptive factors ex vivo that induce neuronal plasticity and may actively diffuse to induce neo-innervation and pain in vivo.
Collapse
Affiliation(s)
- Emerson Krock
- Orthopeadic Research Laboratory, Division of Orthopedic Surgery, McGill University, Montreal, QC, Canada; McGill Scoliosis and Spine Research Group, Montreal, QC, Canada
| | | | | | | | | | | | | | | |
Collapse
|
12
|
JIANG HONG, LI JICHANG, ZHOU TIEZHONG, WANG CHUNHUA, ZHANG HUA, WANG HONGJUN. Colistin-induced apoptosis in PC12 cells: Involvement of the mitochondrial apoptotic and death receptor pathways. Int J Mol Med 2014; 33:1298-304. [DOI: 10.3892/ijmm.2014.1684] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2013] [Accepted: 02/28/2014] [Indexed: 11/06/2022] Open
|
13
|
Gawri R, Rosenzweig DH, Krock E, Ouellet JA, Stone LS, Quinn TM, Haglund L. High mechanical strain of primary intervertebral disc cells promotes secretion of inflammatory factors associated with disc degeneration and pain. Arthritis Res Ther 2014; 16:R21. [PMID: 24457003 PMCID: PMC3979109 DOI: 10.1186/ar4449] [Citation(s) in RCA: 112] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2013] [Accepted: 01/13/2014] [Indexed: 02/02/2023] Open
Abstract
Introduction Excessive mechanical loading of intervertebral discs (IVDs) is thought to alter matrix properties and influence disc cell metabolism, contributing to degenerative disc disease and development of discogenic pain. However, little is known about how mechanical strain induces these changes. This study investigated the cellular and molecular changes as well as which inflammatory receptors and cytokines were upregulated in human intervertebral disc cells exposed to high mechanical strain (HMS) at low frequency. The impact of these metabolic changes on neuronal differentiation was also explored to determine a role in the development of disc degeneration and discogenic pain. Methods Isolated human annulus fibrosus (AF) and nucleus pulposus (NP) cells were exposed to HMS (20% cyclical stretch at 0.001 Hz) on high-extension silicone rubber dishes coupled to a mechanical stretching apparatus and compared to static control cultures. Gene expression of Toll-like receptors (TLRs), neuronal growth factor (NGF) and tumour necrosis factor α (TNFα) was assessed. Collected conditioned media were analysed for cytokine content and applied to rat pheocromocytoma PC12 cells for neuronal differentiation assessment. Results HMS caused upregulation of TLR2, TLR4, NGF and TNFα gene expression in IVD cells. Medium from HMS cultures contained elevated levels of growth-related oncogene, interleukin 6 (IL-6), IL-8, IL-15, monocyte chemoattractant protein 1 (MCP-1), MCP-3, monokine induced by γ interferon, transforming growth factor β1, TNFα and NGF. Exposure of PC12 cells to HMS-conditioned media resulted in both increased neurite sprouting and cell death. Conclusions HMS culture of IVD cells in vitro drives cytokine and inflammatory responses associated with degenerative disc disease and low-back pain. This study provides evidence for a direct link between cellular strain, secretory factors, neoinnervation and potential degeneration and discogenic pain in vivo.
Collapse
|
14
|
Manecka DL, Mahmood SF, Grumolato L, Lihrmann I, Anouar Y. Pituitary adenylate cyclase-activating polypeptide (PACAP) promotes both survival and neuritogenesis in PC12 cells through activation of nuclear factor κB (NF-κB) pathway: involvement of extracellular signal-regulated kinase (ERK), calcium, and c-REL. J Biol Chem 2013; 288:14936-48. [PMID: 23564451 DOI: 10.1074/jbc.m112.434597] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
The pituitary adenylate cyclase-activating polypeptide (PACAP) is a trophic factor that promotes neuronal survival and neurite outgrowth. However, the signaling pathways and the transcriptional mechanisms involved are not completely elucidated. Our previous studies aimed at characterizing the transcriptome of PACAP-differentiated PC12 cells revealed an increase in the expression of nuclear factor κB2 (NF-κB2) gene coding for p100/p52 subunit of NF-κB transcription factor. Here, we examined the role of the NF-κB pathway in neuronal differentiation promoted by PACAP. We first showed that PACAP-driven survival and neuritic extension in PC12 cells are inhibited following NF-κB pathway blockade. PACAP stimulated both c-Rel and p52 NF-κB subunit gene expression and nuclear translocation, whereas c-Rel down-regulation inhibited cell survival and neuritogenesis elicited by the neuropeptide. PACAP-induced c-Rel nuclear translocation was inhibited by ERK1/2 and Ca(2+) blockers. Furthermore, the neuropeptide stimulated NF-κB p100 subunit processing into p52, indicative of activation of the NF-κB alternative pathway. Taken together, our data show that PACAP promotes both survival and neuritogenesis in PC12 cells by activating NF-κB pathway, most likely via classical and alternative signaling cascades involving ERK1/2 kinases, Ca(2+), and c-Rel/p52 dimers.
Collapse
Affiliation(s)
- Destiny-Love Manecka
- INSERM, U982, Laboratory of Neuronal and Neuroendocrine Differentiation and Communication, Institute for Research and Innovation in Biomedicine, University of Rouen, 76821 Mont-Saint-Aignan, France
| | | | | | | | | |
Collapse
|
15
|
Fukumitsu H, Soumiya H, Furukawa S. Knockdown of pre-mRNA cleavage factor Im 25 kDa promotes neurite outgrowth. Biochem Biophys Res Commun 2012; 425:848-53. [PMID: 22898046 DOI: 10.1016/j.bbrc.2012.07.164] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2012] [Accepted: 07/31/2012] [Indexed: 01/16/2023]
Abstract
Mammalian precursor mRNA (pre-mRNA) cleavage factor I (CFIm) plays important roles in the selection of poly(A) sites in a 3'-untranslated region (3'-UTR), producing mRNAs with variable 3' ends. Because 3'-UTRs often contain cis elements that impact stability or localization of mRNA or translation, alternative polyadenylation diversifies utilization of primary transcripts in mammalian cells. However, the physiological role of CFIm remains unclear. CFIm acts as a heterodimer comprising a 25kDa subunit (CFIm25) and one of the three large subunits-CFIm59, CFIm68, or CFIm72. CFIm25 binds directly to RNA and introduces and anchors the larger subunit. To examine the physiological roles of CFIm, we knocked down the CFIm25 gene in neuronal cells using RNA interference. Knockdown of CFIm25 increased the number of primary dendrites of developing hippocampal neurons and promoted nerve growth factor (NGF)-induced neurite extension from rat pheochromocytoma PC12 cells without affecting the morphology of proliferating PC12 cells. On the other hand, CFIm25 knockdown did not influence constitutively active or dominantly negative RhoA suppression or promotion of NGF-induced neurite extension from PC12 cells, respectively. Taken together, our results indicate that endogenous CFIm may promote neuritogenesis in developing neurons by coordinating events upstream of NGF-induced RhoA inactivation.
Collapse
Affiliation(s)
- Hidefumi Fukumitsu
- Laboratory of Molecular Biology, Department of Biofunctional Analysis, Gifu Pharmaceutical University, Daigakunishi 1-25-4, Gifu 501 1196, Japan.
| | | | | |
Collapse
|
16
|
Nabiuni M, Rasouli J, Parivar K, Kochesfehani HM, Irian S, Miyan JA. In vitro effects of fetal rat cerebrospinal fluid on viability and neuronal differentiation of PC12 cells. Fluids Barriers CNS 2012; 9:8. [PMID: 22494846 PMCID: PMC3386012 DOI: 10.1186/2045-8118-9-8] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2011] [Accepted: 04/11/2012] [Indexed: 12/22/2022] Open
Abstract
Background Fetal cerebrospinal fluid (CSF) contains many neurotrophic and growth factors and has been shown to be capable of supporting viability, proliferation and differentiation of primary cortical progenitor cells. Rat pheochromocytoma PC12 cells have been widely used as an in vitro model of neuronal differentiation since they differentiate into sympathetic neuron-like cells in response to growth factors. This study aimed to establish whether PC12 cells were responsive to fetal CSF and therefore whether they might be used to investigate CSF physiology in a stable cell line lacking the time-specific response patterns of primary cells previously described. Methods In vitro assays of viability, proliferation and differentiation were carried out after incubation of PC12 cells in media with and without addition of fetal rat CSF. An MTT tetrazolium assay was used to assess cell viability and/or cell proliferation. Expression of neural differentiation markers (MAP-2 and β-III tubulin) was determined by immunocytochemistry. Formation and growth of neurites was measured by image analysis. Results PC12 cells differentiate into neuronal cell types when exposed to bFGF. Viability and cell proliferation of PC12 cells cultured in CSF-supplemented medium from E18 rat fetuses were significantly elevated relative to the control group. Neuronal-like outgrowths from cells appeared following the application of bFGF or CSF from E17 and E19 fetuses but not E18 or E20 CSF. Beta-III tubulin was expressed in PC12 cells cultured in any media except that supplemented with E18 CSF. MAP-2 expression was found in control cultures and in those with E17 and E19 CSF. MAP2 was located in neurites except in E17 CSF when the whole cell was positive. Conclusions Fetal rat CSF supports viability and stimulates proliferation and neurogenic differentiation of PC12 cells in an age-dependent way, suggesting that CSF composition changes with age. This feature may be important in vivo for the promotion of normal brain development. There were significant differences in the effects on PC12 cells compared to primary cortical cells. This suggests there is an interaction in vivo between developmental stage of cells and the composition of CSF. The data presented here support an important, perhaps driving role for CSF composition, specifically neurotrophic factors, in neuronal survival, proliferation and differentiation. The effects of CSF on PC12 cells can thus be used to further investigate the role of CSF in driving development without the confounding issues of using primary cells.
Collapse
Affiliation(s)
- Mohammad Nabiuni
- Faculty of Life sciences, The University of Manchester, AV Hill Building, Oxford Road, Manchester, M13 9PT, UK.
| | | | | | | | | | | |
Collapse
|
17
|
Raoult E, Roussel BD, Bénard M, Lefebvre T, Ravni A, Ali C, Vivien D, Komuro H, Fournier A, Vaudry H, Vaudry D, Galas L. Pituitary adenylate cyclase-activating polypeptide (PACAP) stimulates the expression and the release of tissue plasminogen activator (tPA) in neuronal cells: involvement of tPA in the neuroprotective effect of PACAP. J Neurochem 2011; 119:920-31. [DOI: 10.1111/j.1471-4159.2011.07486.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
|
18
|
Habauzit D, Flouriot G, Pakdel F, Saligaut C. Effects of estrogens and endocrine-disrupting chemicals on cell differentiation-survival-proliferation in brain: contributions of neuronal cell lines. JOURNAL OF TOXICOLOGY AND ENVIRONMENTAL HEALTH. PART B, CRITICAL REVIEWS 2011; 14:300-327. [PMID: 21790314 DOI: 10.1080/10937404.2011.578554] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
Estrogens and estrogen receptors (ER) are key actors in the control of differentiation and survival and act on extrareproductive tissues such as brain. Thus, estrogens may display neuritogenic effects during development and neuroprotective effects in the pathophysiological context of brain ischemia and neurodegenerative pathologies like Alzheimer's disease or Parkinson's disease. Some of these effects require classical transcriptional "genomic" mechanisms through ER, whereas other effects appear to rely clearly on "membrane-initiated mechanisms" through cytoplasmic signal transduction pathways. Disturbances of these mechanisms by endocrine-disrupting chemicals (EDC) may exert adverse effects on brain. Some EDC may act via ER-independent mechanisms but might cross-react with endogenous estrogen. Other EDC may act through ER-dependent mechanisms and display agonistic/antagonistic estrogenic properties. Because of these potential effects of EDC, it is necessary to establish sensitive cell-based assays to determine EDC effects on brain. In the present review, some effects of estrogens and EDC are described with focus on ER-mediated effects in neuronal cells. Particular attention is given to PC12 cells, an interesting model to study the mechanisms underlying ER-mediated differentiating and neuroprotective effects of estrogens.
Collapse
Affiliation(s)
- Denis Habauzit
- UMR CNRS 6026 (Interactions Cellulaires et Moléculaires, Equipe RED), Université de Rennes 1, Rennes, France
| | | | | | | |
Collapse
|
19
|
Jiao L, Zhang Y, Hu C, Wang YG, Huang A, He C. Rap1GAP interacts with RET and suppresses GDNF-induced neurite outgrowth. Cell Res 2010; 21:327-37. [PMID: 20877310 DOI: 10.1038/cr.2010.139] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Glial cell line-derived neurotrophic factor (GDNF) was originally recognized for its ability to promote survival of midbrain dopaminergic neurons, but it has since been demonstrated to be crucial for the survival and differentiation of many neuronal subpopulations, including motor neurons, sympathetic neurons, sensory neurons and enteric neurons. To identify possible effectors or regulators of GDNF signaling, we performed a yeast two-hybrid screen using the intracellular domain of RET, the common signaling receptor of the GDNF family, as bait. Using this approach, we identified Rap1GAP, a GTPase-activating protein (GAP) for Rap1, as a novel RET-binding protein. Endogenous Rap1GAP co-immunoprecipitated with RET in neural tissues, and RET and Rap1GAP were co-expressed in dopaminergic neurons of the mesencephalon. In addition, overexpression of Rap1GAP attenuated GDNF-induced neurite outgrowth, whereas suppressing the expression of endogenous Rap1GAP by RNAi enhanced neurite outgrowth. Furthermore, using co-immunoprecipitation analyses, we found that the interaction between RET and Rap1GAP was enhanced following GDNF treatment. Mutagenesis analysis revealed that Tyr981 in the intracellular domain of RET was crucial for the interaction with Rap1GAP. Moreover, we found that Rap1GAP negatively regulated GNDF-induced ERK activation and neurite outgrowth. Taken together, our results suggest the involvement of a novel interaction of RET with Rap1GAP in the regulation of GDNF-mediated neurite outgrowth.
Collapse
Affiliation(s)
- Li Jiao
- Institute of Neuroscience and Key Laboratory of Molecular Neurobiology of Ministry of Education, Neuroscience Research Center of Changzheng Hospital, Second Military Medical University, 800 Xiangyin Road, Shanghai 200433, China
| | | | | | | | | | | |
Collapse
|
20
|
Dejda A, Chan P, Seaborn T, Coquet L, Jouenne T, Fournier A, Vaudry H, Vaudry D. Involvement of stathmin 1 in the neurotrophic effects of PACAP in PC12 cells. J Neurochem 2010; 114:1498-510. [PMID: 20569302 DOI: 10.1111/j.1471-4159.2010.06873.x] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Rat pheochromocytoma PC12 cells have been widely used to investigate the neurotrophic activities of pituitary adenylate cyclase-activating polypeptide (PACAP). In particular, PACAP has been shown to promote differentiation and to inhibit apoptosis of PC12 cells. In order to identify the mechanisms mediating these effects, we sought for proteins that are phosphorylated upon PACAP treatment. High-performance liquid chromatography and 2D gel electrophoresis analysis, coupled with mass spectrometry, revealed that stathmin 1 is strongly phosphorylated within only 5 min of exposure to PACAP. Western blot experiments confirmed that PACAP induced a robust phosphorylation of stathmin 1 in a time-dependent manner. On the other hand, PACAP decreased stathmin 1 gene expression. Investigations of the signaling mechanisms known to be activated by PACAP revealed that phosphorylation of stathmin 1 was mainly mediated through the protein kinase A and mitogen-activated protein kinase pathways. Blockage of stathmin 1 expression with small interfering RNA did not affect PC12 cell differentiation induced by PACAP but reduced the ability of the peptide to inhibit caspase 3 activity and significantly decreased its neuroprotective action. Taken together, these data demonstrate that stathmin 1 is involved in the neurotrophic effect of PACAP in PC12 cells.
Collapse
Affiliation(s)
- Agnieszka Dejda
- INSERM U982, Institut Fédératif de Recherches Multidisciplinaires sur les Peptides (IFRMP 23), Université de Rouen, Mont-Saint-Aignan, France
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Huang X, Li Q, Li H, Guo L. Neuroprotective and antioxidative effect of cactus polysaccharides in vivo and in vitro. Cell Mol Neurobiol 2009; 29:1211-21. [PMID: 19517228 DOI: 10.1007/s10571-009-9417-z] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/21/2008] [Accepted: 05/14/2009] [Indexed: 11/29/2022]
Abstract
Cactus polysaccharides (CP), some of the active components in Opuntia dillenii Haw have been reported to display neuroprotective effects in rat brain slices. In the present study, we investigated the neuroprotective properties of CP and their potential mechanisms on brain ischemia-reperfusion injury in rats, and on oxidative stress-induced damage in PC12 cells. Male Sprague-Dawley rats with ischemia following middle cerebral artery occlusion and reperfusion were investigated. CP (200 mg/kg) significantly decreased the neurological deficit score, reduced infarct volume, decreased neuronal loss in cerebral cortex, and remarkably reduced the protein synthesis of inducible nitric oxide synthase which were induced by ischemia and reperfusion. Otherwise, the protective effect of CP was confirmed in in vitro study. CP protected PC12 cells against hydrogen peroxide (H(2)O(2)) insult. Pretreatment with CP prior to H(2)O(2) exposure significantly elevated cell viability, reduced H(2)O(2)-induced apoptosis, and decreased both intracellular and total accumulation of reactive oxygen species (ROS) production. Furthermore, CP also reversed the upregulation of Bax/Bcl-2 mRNA ratio, the downstream cascade following ROS. These results suggest that CP may be a candidate compound for the treatment of ischemia and oxidative stress-induced neurodegenerative disease.
Collapse
Affiliation(s)
- Xianju Huang
- Department of Pharmacology, Tongji Medical College, Huazhong University of Science and Technology, Hangkong Road 13#, 430030, Wuhan, People's Republic of China.
| | | | | | | |
Collapse
|
22
|
Vaudry D, Falluel-Morel A, Bourgault S, Basille M, Burel D, Wurtz O, Fournier A, Chow BKC, Hashimoto H, Galas L, Vaudry H. Pituitary Adenylate Cyclase-Activating Polypeptide and Its Receptors: 20 Years after the Discovery. Pharmacol Rev 2009; 61:283-357. [DOI: 10.1124/pr.109.001370] [Citation(s) in RCA: 829] [Impact Index Per Article: 55.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
|
23
|
Fila T, Trazzi S, Crochemore C, Bartesaghi R, Ciani E. Lot1 is a key element of the pituitary adenylate cyclase-activating polypeptide (PACAP)/cyclic AMP pathway that negatively regulates neuronal precursor proliferation. J Biol Chem 2009; 284:15325-38. [PMID: 19346254 DOI: 10.1074/jbc.m109.002329] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
The tumor suppressor gene Lot1 is highly expressed during brain development. During cerebellar development, Lot1 is expressed by proliferating granule cells with a time course matching the expression of the pituitary adenylate cyclase-activating polypeptide (PACAP) receptor, a neuropeptide receptor that plays an important role in the regulation of granule cell proliferation/survival. Although it has become clear that Lot1 is a negative regulator of cell division in tumor cells, its role in neuronal proliferation is not understood. We previously demonstrated that in cerebellar granule cells Lot1 expression is regulated by the PACAP/cAMP system. The aim of this study was to investigate the role played by Lot1 in neuron proliferation/survival and to identify the molecular mechanisms underlying its actions. Using a Lot1-inducible expression system, we found that in PC12 cells Lot1 negatively regulates proliferation and favors differentiation by up-regulating the expression of the PACAP receptor. In cerebellar granule cells in culture, an increase in Lot1 expression was paralleled by inhibition of proliferation and up-regulation of the PACAP receptor, which in turn positively regulated Lot1 expression. Silencing of Lot1 leads to an increase in granule cell proliferation and a reduction in survival. Confirming the in vitro results, in vivo experiments showed that PACAP induced an increase in Lot1 expression that was paralleled by inhibition of cerebellar granule cell proliferation. These data show that Lot1 is a key element of the PACAP/cAMP pathway that negatively regulates neuronal precursor proliferation. The existence of a PACAP receptor/Lot1-positive feedback loop may powerfully regulate neural proliferation during critical phases of cerebellar development.
Collapse
Affiliation(s)
- Tatiana Fila
- Department of Human and General Physiology, University of Bologna, Piazza di Porta San Donato 2, Bologna, Italy
| | | | | | | | | |
Collapse
|
24
|
Krug AW, Langbein H, Ziegler CG, Bornstein SR, Eisenhofer G, Ehrhart-Bornstein M. Dehydroepiandrosterone-sulphate (DHEA-S) promotes neuroendocrine differentiation of chromaffin pheochromocytoma PC12 cells. Mol Cell Endocrinol 2009; 300:126-31. [PMID: 19022340 DOI: 10.1016/j.mce.2008.10.026] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/14/2008] [Revised: 10/16/2008] [Accepted: 10/17/2008] [Indexed: 02/05/2023]
Abstract
The major source for dehydroepiandrosterone (DHEA) and its sulphate compound DHEA-S is the inner zone of the adrenal cortex, which is in direct contact to adrenomedullary chromaffin cells. Due to their close proximity, direct interactions of DHEA and DHEA-S with chromaffin cells during adrenal gland development and throughout the whole life span are hypothesized. A possible direct effect of DHEA-S and the cellular and molecular mechanisms of DHEA-S action on chromaffin cells remain unresolved. Therefore, in this study, we aimed at clarifying DHEA-S effects and mechanisms of action on rat chromaffin PC12 cells. DHEA-S (10(-6)mol/l) inhibited nerve growth factor (NGF, 20ng/ml)-induced cell proliferation by 66% (n=4, p<0.001). In NGF-stimulated cells, neuronal differentiation was inhibited by DHEA-S, as demonstrated by a 22% reduction (n=3; p<0.05) of neuronal differentiation marker expression, synaptosome-associated protein of 25kDa (SNAP-25), and a 59% (n=6; p<0.001) decrease in neurite outgrowth. Moreover, DHEA-S stimulated expression of endocrine marker chromogranin A (CgA) by 31% (n=4; p<0.05 vs. control) and catecholamine release from NGF-treated PC12 cells by 229% (n=3-5; p<0.001), indicating a DHEA-S-induced shift towards neuroendocrine differentiation. On a molecular level, DHEA-S diminished NGF-induced ERK1/2 phosphorylation. Taken together, DHEA-S inhibited NGF-induced proliferation and neuronal differentiation and shifted cells towards a more endocrine phenotype. Interference of DHEA-S with NGF-stimulated ERK1/2 activation might be involved in this effect. Our study provides support for the notion that adrenocortical-derived DHEA-S impacts adrenomedullary chromaffin cells during development and differentiation.
Collapse
Affiliation(s)
- Alexander W Krug
- Carl Gustav Carus University Hospital, Medical Clinic III, University of Dresden, Germany.
| | | | | | | | | | | |
Collapse
|
25
|
Kawakami M, Inagawa R, Hosokawa T, Saito T, Kurasaki M. Mechanism of apoptosis induced by copper in PC12 cells. Food Chem Toxicol 2008; 46:2157-64. [DOI: 10.1016/j.fct.2008.02.014] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2007] [Revised: 02/01/2008] [Accepted: 02/19/2008] [Indexed: 02/03/2023]
|
26
|
Zhang J, Chen X. DeltaNp73 modulates nerve growth factor-mediated neuronal differentiation through repression of TrkA. Mol Cell Biol 2007; 27:3868-80. [PMID: 17353261 PMCID: PMC1899982 DOI: 10.1128/mcb.02112-06] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
p73, a member of the p53 family, expresses two classes of proteins: the full-length TAp73 and the N-terminally truncated DeltaNp73. While TAp73 possesses many p53-like features, DeltaNp73 is dominant negative towards TAp73 and p53 and appears to have distinct functions in tumorigenesis and neuronal development. Given its biological importance, we investigated the role of DeltaNp73 in nerve growth factor (NGF)-mediated neuronal differentiation in PC12 cells. We show that overexpression of DeltaNp73alpha or DeltaNp73beta inhibits NGF-mediated neuronal differentiation in both p53-dependent and -independent manners. In line with this, we showed that the level of endogenous DeltaNp73 is progressively diminished in differentiating PC12 cells upon NGF treatment and knockdown of DeltaNp73 promotes NGF-mediated neuronal differentiation. Interestingly, we found that the ability of DeltaNp73 to suppress NGF-mediated neuronal differentiation is correlated with its ability to regulate the expression of TrkA, the high-affinity NGF receptor. Specifically, we found that DeltaNp73 directly binds to the TrkA promoter and transcriptionally represses TrkA expression, which in turn attenuates the NGF-mediated mitogen-activated protein kinase pathway. Conversely, the steady-state level of TrkA is increased upon knockdown of DeltaNp73. Furthermore, we found that histone deacetylase 1 (HDAC1) and HDAC2 are recruited by DeltaNp73 to the TrkA promoter and act as corepressors to suppress TrkA expression, which can be relieved by trichostatin A, an HDAC inhibitor. Taken together, we conclude that DeltaNp73 negatively regulates NGF-mediated neuronal differentiation by transrepressing TrkA.
Collapse
Affiliation(s)
- Jin Zhang
- Center for Comparative Oncology, 2128 Tupper Hall, University of California at Davis, Davis, CA 95616, USA
| | | |
Collapse
|
27
|
The effects of PACAP on neural cell proliferation. ACTA ACUST UNITED AC 2006; 137:50-7. [PMID: 17011642 DOI: 10.1016/j.regpep.2006.03.009] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2006] [Revised: 03/20/2006] [Accepted: 03/30/2006] [Indexed: 01/25/2023]
Abstract
PACAP and its receptors are expressed in growth zones of the brain. By stimulating PAC(1)-receptors PACAP can enhance, as well as reduce, the proliferation rate in a cell-type dependent manner. PACAP can enhance the proliferation rate by activating phospholipase C and protein kinase C, although other signal transduction pathways may also be responsible. PACAP can suppress proliferation by inhibiting protein complexes of the cyclins D and E with the cyclin-dependent kinases 4/6 and 2, respectively, which are necessary for entry into the cell cycle. PACAP seems to exert these inhibitory effects by acting via the Sonic hedgehog glycoprotein and the small GTPase RhoA. Also, the activation of a cyclin-dependent kinase inhibitor has been suggested. The signal transduction pathways mediating the effects of PACAP on proliferation are discussed.
Collapse
|
28
|
Ravni A, Bourgault S, Lebon A, Chan P, Galas L, Fournier A, Vaudry H, Gonzalez B, Eiden LE, Vaudry D. The neurotrophic effects of PACAP in PC12 cells: control by multiple transduction pathways. J Neurochem 2006; 98:321-9. [PMID: 16805827 DOI: 10.1111/j.1471-4159.2006.03884.x] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Pituitary adenylate cyclase-activating polypeptide (PACAP) and vasoactive intestinal polypeptide (VIP) are closely related members of the secretin superfamily of neuropeptides expressed in both the brain and peripheral nervous system, and they exhibit neurotrophic and neurodevelopmental effects in vivo. Like the index member of the Trk receptor ligand family, nerve growth factor (NGF), PACAP promotes the differentiation of PC12 cells, a well-established cell culture model, to investigate neuronal differentiation, survival and function. Stimulation of catecholamine secretion and enhanced neuropeptide biosynthesis are effects exerted by PACAP at the adrenomedullary synapse in vivo and on PC12 cells in vitro through stimulation of the specific PAC1 receptor. Induction of neuritogenesis, growth arrest, and promotion of cell survival are effects of PACAP that occur in developing cerebellar, hippocampal and cortical neurons, as well as in the more tractable PC12 cell model. Study of the mechanisms through which PACAP exerts its various effects on cell growth, morphology, gene expression and survival, i.e. its actions as a neurotrophin, in PC12 cells is the subject of this review. The study of neurotrophic signalling by PACAP in PC12 cells reveals that multiple independent pathways are coordinated in the PACAP response, some activated by classical and some by novel or combinatorial signalling mechanisms.
Collapse
Affiliation(s)
- Aurélia Ravni
- Laboratory of Cellular and Molecular Neuroendocrinology, European Institute for Peptide Research, University of Rouen, Mont-Saint-Aignan, France
| | | | | | | | | | | | | | | | | | | |
Collapse
|
29
|
Yang W, Liu P, Liu Y, Wang Q, Tong Y, Ji J. Proteomic analysis of rat pheochromocytoma PC12 cells. Proteomics 2006; 6:2982-90. [PMID: 16622837 DOI: 10.1002/pmic.200500036] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
PC12 cell line is well documented and widely applied as many kinds of models in neurobiological and neurochemical studies. Yet a thorough proteomic analysis has not been performed so far. Here we report the construction of a large-scale 2-D protein database for PC12 cells. The proteins extracted from PC12 cells were separated by 2-DE and identified by MALDI-TOF/TOF MS. A total of 1080 protein spots, excised from three different 2-D gels, were identified with high confidence. These proteins represent 474 different gene products, mainly binding proteins and enzymes. Three hundred and seven identified protein spots were located in the low-molecular weight region below 20 kDa. This database today represents one of the largest 2-D databases for higher eukaryotic cell proteomes and for low-molecular weight proteins. In addition, fragment ion spectra obtained by TOF/TOF confirmed that calcylin in PC12 cells was N-acetylated. The database of PC12 proteome is expected to be a powerful tool for neuroscientists.
Collapse
Affiliation(s)
- Wei Yang
- Department of Biochemistry and Molecular Biology, College of Life Sciences, National Laboratory of Protein Engineering and Plant Genetic Engineering, Peking University, Beijing, P. R. China
| | | | | | | | | | | |
Collapse
|
30
|
Uwabe KI, Matsumoto M, Nagata K. Monokine Induced by Interferon-γ Acts as a Neurotrophic Factor on PC12 Cells and Rat Primary Sympathetic Neurons. J Biol Chem 2005; 280:34268-77. [PMID: 16055446 DOI: 10.1074/jbc.m502667200] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
We found that a monokine induced by interferon-gamma (Mig, CXCL9), which belongs to the CXC chemokine subfamily, acts as a neurotrophic factor on PC12 cells and rat primary sympathetic neurons. PC12 cells were shown to express a single class of high affinity binding sites for Mig (670 receptors/cell, Kd = 2.9 nm). Mig induced neurite outgrowth in PC12 cells in a dose-dependent manner. Comparison of extracellular signal-regulated kinase signaling pathways between Mig and nerve growth factor (NGF) revealed that these pathways are crucial for Mig action as well as NGF. K252a, an inhibitor of tyrosine autophosphorylation of tyrosine kinase receptors (Trks) did not inhibit the action of Mig, suggesting that Mig action occurs via a different receptor from that of NGF. Furthermore, Mig as well as NGF promoted PC12 survival under serum-free conditions and activated Akt/protein kinase B downstream from phosphatidylinositol 3-kinase (PI3K). Because the PI3K inhibitor LY294002 prevented the Mig- and NGF-induced survival effect, this effect is probably mediated by the PI3K signaling pathway. Mig also promoted survival of rat primary sympathetic neurons that die when deprived of NGF. These results suggest that chemokines, including Mig (CXCL9) have neurotrophic effects on the nervous system.
Collapse
Affiliation(s)
- Ken-Ichiro Uwabe
- Discovery Research Laboratories, Shionogi & Co., Ltd., 12-4, Sagisu 5-chome, Fukushima-ku, Osaka 553-0002, Japan.
| | | | | |
Collapse
|
31
|
Bau C, Middlemiss PJ, Hindley S, Jiang S, Ciccarelli R, Caciagli F, Diiorio P, Werstiuk ES, Rathbone MP. Guanosine stimulates neurite outgrowth in PC12 cells via activation of heme oxygenase and cyclic GMP. Purinergic Signal 2005; 1:161-72. [PMID: 18404501 PMCID: PMC2096532 DOI: 10.1007/s11302-005-6214-0] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2004] [Accepted: 10/22/2004] [Indexed: 12/19/2022] Open
Abstract
Undifferentiated rat pheochromocytoma (PC12) cells extend neurites when cultured in the presence of nerve growth factor (NGF). Extracellular guanosine synergistically enhances NGF-dependent neurite outgrowth. We investigated the mechanism by which guanosine enhances NGF-dependent neurite outgrowth. Guanosine administration to PC12 cells significantly increased guanosine 3-5-cyclic monophosphate (cGMP) within the first 24 h whereas addition of soluble guanylate cyclase (sGC) inhibitors abolished guanosine-induced enhancement of NGF-dependent neurite outgrowth. sGC may be activated either by nitric oxide (NO) or by carbon monoxide (CO). \documentclass[12pt]{minimal}
\usepackage{amsmath}
\usepackage{wasysym}
\usepackage{amsfonts}
\usepackage{amssymb}
\usepackage{amsbsy}
\usepackage{mathrsfs}
\usepackage{upgreek}
\setlength{\oddsidemargin}{-69pt}
\begin{document}
$$N^{\omega } $$
\end{document}-Nitro-l-arginine methyl ester (l-NAME), a non-isozyme selective inhibitor of nitric oxide synthase (NOS), had no effect on neurite outgrowth induced by guanosine. Neither nNOS (the constitutive isoform), nor iNOS (the inducible isoform) were expressed in undifferentiated PC12 cells, or under these treatment conditions. These data imply that NO does not mediate the neuritogenic effect of guanosine. Zinc protoporphyrin-IX, an inhibitor of heme oxygenase (HO), reduced guanosine-dependent neurite outgrowth but did not attenuate the effect of NGF. The addition of guanosine plus NGF significantly increased the expression of HO-1, the inducible isozyme of HO, after 12 h. These data demonstrate that guanosine enhances NGF-dependent neurite outgrowth by first activating the constitutive isozyme HO-2, and then by inducing the expression of HO-1, the enzymes responsible for CO synthesis, thus stimulating sGC and increasing intracellular cGMP.
Collapse
Affiliation(s)
- Christian Bau
- Department of Medicine, McMaster University, Health Sciences Centre, Hamilton, Ontario, Canada
| | | | | | | | | | | | | | | | | |
Collapse
|
32
|
Falluel-Morel A, Aubert N, Vaudry D, Basille M, Fontaine M, Fournier A, Vaudry H, Gonzalez BJ. Opposite regulation of the mitochondrial apoptotic pathway by C2-ceramide and PACAP through a MAP-kinase-dependent mechanism in cerebellar granule cells. J Neurochem 2005; 91:1231-43. [PMID: 15569266 DOI: 10.1111/j.1471-4159.2004.02810.x] [Citation(s) in RCA: 84] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023]
Abstract
The sphingomyelin-derived messenger ceramides provoke neuronal apoptosis through caspase-3 activation, while the neuropeptide pituitary adenylate cyclase-activating polypeptide (PACAP) promotes neuronal survival and inhibits caspase-3 activity. However, the mechanisms leading to the opposite regulation of caspase-3 by C2-ceramide and PACAP are currently unknown. Here, we show that PACAP prevents C2-ceramide-induced inhibition of mitochondrial potential and C2-ceramide-evoked cytochrome c release. C2-ceramide stimulated Bax expression, but had no effect on Bcl-2, while PACAP abrogated the action of C2-ceramide on Bax and stimulated Bcl-2 expression. The effects of C2-ceramide and PACAP on Bax and Bcl-2 were blocked, respectively, by the JNK inhibitor L-JNKI1 and the MEK inhibitor U0126. L-JNKI1 prevented the alteration of mitochondria induced by C2-ceramide while U0126 suppressed the protective effect of PACAP against the deleterious action of C2-ceramide on mitochondrial potential. Moreover, L-JNKI1 inhibited the stimulatory effect of C2-ceramide on caspase-9 and -3 and prevented C2-ceramide-induced cell death. U0126 blocked PACAP-induced Bcl-2 expression, abrogated the inhibitory effect of PACAP on ceramide-induced caspase-9 activity, and promoted granule cell death. The present study reveals that C2-ceramide and PACAP exert opposite effects on Bax and Bcl-2 through, respectively, JNK- and ERK-dependent mechanisms. These data indicate that the mitochondrial pathway plays a pivotal role in the pro- and anti-apoptotic effects of C2-ceramide and PACAP.
Collapse
Affiliation(s)
- Anthony Falluel-Morel
- European Institute for Peptide Research (IFRMP 23), Laboratory of Cellular and Molecular Neuroendocrinology, University of Rouen, Mont-Saint-Aignan, France
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Topalli I, Etgen AM. Insulin-like growth factor-I receptor and estrogen receptor crosstalk mediates hormone-induced neurite outgrowth in PC12 cells. Brain Res 2005; 1030:116-24. [PMID: 15567343 DOI: 10.1016/j.brainres.2004.09.057] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/27/2004] [Indexed: 01/08/2023]
Abstract
Estradiol (E(2)) and insulin-like growth factor-I (IGF-I) can act independently or in concert to promote neurite outgrowth in vivo and in cultured neurons. This study examined the role of crosstalk between estrogen receptor (ER)alpha and the IGF-I receptor as a critical mediator of hormone- and growth factor-dependent neurite outgrowth in a homogenous cell system. We used control PC12 cells and PC12 cells stably transfected with ER alpha, both of which express IGF-I receptor. Cells were treated for 1 week with vehicle, 1 nM E(2) or 100 ng/ml IGF-I alone or with E(2) or IGF-I in the presence of either the IGF-I receptor antagonist JB1 or the ER antagonist ICI 182,780. IGF-I significantly increased neurite outgrowth, as measured by the percentage of process-bearing cells, and absolute neurite length per cell in both control and ER alpha-transfected PC12 cells. In contrast, E(2) increased process formation and extension only in PC12 cells that were stably transfected with ER alpha. ICI 182,780 and JB1 blocked the IGF-I-induced increases in neurite length in both cell types. The efficacy of ICI 182,780 in control PC12 cells may have been due to the upregulation of ER alpha in these cells by the 7-day treatment with IGF-I. The ER and IGF-I receptor antagonists similarly blocked the E(2)-induced increase in neurite lengths in ER alpha-transfected cells. Immunofluorescent analysis of the cellular distribution of an axonal marker, phospho-neurofilament, verified that the processes extended by PC12 cells were neurites. These data suggest that receptor crosstalk between IGF-I receptors and ER alpha has an important role in neurite formation and extension even in a single-cell system.
Collapse
Affiliation(s)
- Ilir Topalli
- Department of Neuroscience, Forchheimer 113, Albert Einstein College of Medicine, Bronx, NY 10461, USA
| | | |
Collapse
|
34
|
Taira E, Kohama K, Tsukamoto Y, Okumura S, Miki N. Gicerin/CD146 is involved in neurite extension of NGF-treated PC12 cells. J Cell Physiol 2005; 204:632-7. [PMID: 15880440 DOI: 10.1002/jcp.20365] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Gicerin/CD146 is a cell adhesion molecule, which belongs to the immunoglobulin (Ig) superfamily. We have reported that it has a homophilic binding activity, which participates in the neurite extension from embryonic neurons. To elucidate how gicerin is involved in the neurite extension mechanism, we employed PC12 cells, which expresses gicerin/CD146. PC12 cells extend longer neurites by nerve growth factor (NGF) on gicerin substrate than on without gicerin substrate, which indicates that gicerin participates in neurite extension by NGF. We also found that the expression of gicerin in PC12 cells is induced by NGF. Over-expression of gicerin also promotes neurite extension by gicerin-gicerin homophilic interaction. These findings suggested that increase of gicerin expression by NGF promotes the gicerin-gicerin homophilic interaction resulting in the neurite extension.
Collapse
Affiliation(s)
- Eiichi Taira
- Department of Pharmacology, Iwate Medical School, Uchimaru, Morioka, Iwate, Japan.
| | | | | | | | | |
Collapse
|
35
|
Schmitt JM, Wayman GA, Nozaki N, Soderling TR. Calcium Activation of ERK Mediated by Calmodulin Kinase I. J Biol Chem 2004; 279:24064-72. [PMID: 15150258 DOI: 10.1074/jbc.m401501200] [Citation(s) in RCA: 127] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Elevated intracellular Ca(2+) triggers numerous signaling pathways including protein kinases such as the calmodulin-dependent kinases (CaMKs) and the extracellular signal-regulated kinases (ERKs). In the present study we examined Ca(2+)-dependent "cross-talk" between these two protein kinase families. Using a combination of pharmacological inhibitors and dominant-negative kinases (dnKinase), we identified a requirement for CaMKK acting through CaMKI in the stimulation of ERKs upon depolarization of the neuroblastoma cell line, NG108. Depolarization stimulated prolonged ERK and JNK activation that was blocked by the CaMKK inhibitor, STO-609; this inhibition of ERK activation by STO-609 was rescued by expression of a STO-609-insensitive mutant of CaMKK. However, activation of ERK by epidermal growth factor or carbachol were not suppressed by inhibition of CaMKK, indicating specificity for this "cross-talk." To identify the downstream target of CaMKK that mediated ERK activation upon depolarization, dnKinases were expressed. The dnCaMKI completely suppressed ERK2 activation whereas dnAKT/PKB or nuclear-targeted dnCaMKIV, other substrates for CaMKK, were not inhibitory. ERK activation upon depolarization or transfection with constitutively active (ca) CaMKI was blocked by dnRas. Additionally, depolarization of NG108 cells promoted neurite outgrowth, and this effect was blocked by inhibition of either CaMKK (STO-609) or ERK (UO126). Co-transfection with caCaMKK plus caCaMKI also stimulated neurite outgrowth that was blocked by inhibition of ERK (UO126). These data are the first to suggest that ERK activation and neurite outgrowth in response to depolarization are mediated by CaMKK activation of CaMKI.
Collapse
Affiliation(s)
- John M Schmitt
- Vollum Institute, Oregon Health and Sciences University, Portland 97239, USA
| | | | | | | |
Collapse
|
36
|
Pettersson LME, Heine T, Verge VMK, Sundler F, Danielsen N. PACAP mRNA is expressed in rat spinal cord neurons. J Comp Neurol 2004; 471:85-96. [PMID: 14983478 DOI: 10.1002/cne.20015] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
This study examines the expression of pituitary adenylate cyclase activating polypeptide (PACAP) mRNA in the rat spinal cord during normal conditions and in response to sciatic nerve transection. Previously, PACAP immunoreactivity has been found in fibers in the spinal cord dorsal horn and around the central canal and in neurons in the intermediolateral column (IML). Furthermore, in the dorsal root ganglia, PACAP immunoreactivity and PACAP mRNA expression have been observed preferentially in nerve cell bodies of smaller diameter terminating in the superficial laminae of the dorsal horn. However, neuronal expression of PACAP mRNA in adult rat spinal cord appeared limited to neurons of the IML. By using a refined in situ hybridization protocol, we now detect PACAP mRNA expression in neurons primarily in laminae I and II, but also in deeper laminae of the spinal cord dorsal horn and around the central canal. In addition, PACAP mRNA expression is observed in a few neurons in the ventral horn. PACAP expression in the ventral horn is increased in a population of large neurons, most likely motor neurons, both after distal and proximal sciatic nerve transection. The proposed role of PACAP in nociception is strengthened by our findings of PACAP mRNA-expressing neurons in the superficial laminae of the dorsal horn. Furthermore, increased expression of PACAP in ventral horn neurons, in response to nerve transection, suggests a role for PACAP in repair/regeneration of motor neurons.
Collapse
Affiliation(s)
- Lina M E Pettersson
- Department of Physiological Sciences, Section for Neuroendocrine Cell Biology, Lund University, SE-221 84 Lund, Sweden.
| | | | | | | | | |
Collapse
|
37
|
Vaudry D, Taupenot L. Fast-breaking results on the PACAP/VIP/secretin peptide family in chromaffin cells. Ann N Y Acad Sci 2002; 971:460-6. [PMID: 12438165 DOI: 10.1111/j.1749-6632.2002.tb04509.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
The neuropeptide pituitary adenylyl cyclase-activating polypeptide (PACAP) has been reported to be a potent regulator of chromaffin cell activity. In particular, PACAP stimulates catecholamine biosynthesis as well as the expression of various genes, including chromogranin A, neuropeptide Y, enkephalins, vasoactive intestinal polypeptide, and PACAP itself. The mechanisms involved in the effects of PACAP on chromaffin cells have been investigated using rat pheochromocytoma PC12 cells. This cell line turned out to be a suitable model in which to study the neurotrophic activities of PACAP. Recent studies using transgenic mice have shown that in the sympathoadrenal system, PACAP acts as an "emergency response" cotransmitter involved in the regulation of insulin-induced hypoglycemia.
Collapse
Affiliation(s)
- David Vaudry
- Laboratory of Cellular and Molecular Regulation, National Institute of Mental Health, Bethesda, Maryland 20892, USA.
| | | |
Collapse
|