1
|
Seidl LF, Winter S, Aigner L, Schartner J. A Neurofilament-L reporter cell line for the quantification of early neuronal differentiation: A Bioassay for neurotrophic activities. Heliyon 2024; 10:e24753. [PMID: 38304771 PMCID: PMC10831791 DOI: 10.1016/j.heliyon.2024.e24753] [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: 03/15/2023] [Revised: 12/12/2023] [Accepted: 01/12/2024] [Indexed: 02/03/2024] Open
Abstract
Background Neurotrophic activity constitutes a crucial factor in the recovery from neurological injuries and is impaired in neurodegenerative disorders. Preclinical studies of neurotrophic factors to improve outcome of neurodegenerative diseases have yielded promising results. However, due to the complexity of these therapies, the clinical translation of this approach was so far not successful and more feasible treatments with neurotrophic activity may be promising alternatives. Therefore, highly sensitive and robust assays for compound screening are required. New method Nerve growth factor is known to induce Neurofilament-L (NF-L) expression in a rat pheochromocytoma cell line (PC12 cells) during early neuronal differentiation. We generated and characterized an enhanced green fluorescent protein (EGFP)-NF-L reporter PC12 cell line for the development of a cell-based assay (designated Neurofilament-L Bioassay) that allows straightforward quantification of early neuronal differentiation based on NF-L expression. Results Using Cerebrolysin® as a role model for a pharmacological compound that stimulates neurotrophic activity in the central nervous system, the Neurofilament-L Bioassay was proved to be a robust, specific, and reproducible method. Comparison with existing methods It was already shown that NF-L expression correlates with neurite outgrowth in PC12 cells. Currently, quantification of neurite outgrowth is the most commonly used method to evaluate neuronal differentiation in PC12 cells, an approach that is time-consuming and of high variability. Conclusions This work describes the development of an EGFP-NF-L reporter PC12 cell-based assay as a robust and reproducible tool for "high throughput" compound screening for neurotrophic activity.
Collapse
Affiliation(s)
- Lisa-Franziska Seidl
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
- EVER Neuro Pharma, Oberburgau 3, 4866 Unterach, Austria
| | - Stefan Winter
- EVER Neuro Pharma, Oberburgau 3, 4866 Unterach, Austria
| | - Ludwig Aigner
- Institute of Molecular Regenerative Medicine, Paracelsus Medical University, Salzburg, Austria
| | | |
Collapse
|
2
|
Gao AX, Xia TC, Lin LS, Dong TT, Tsim KW. The neurotrophic activities of brain-derived neurotrophic factor are potentiated by binding with apigenin, a common flavone in vegetables, in stimulating the receptor signaling. CNS Neurosci Ther 2023; 29:2787-2799. [PMID: 37101380 PMCID: PMC10493664 DOI: 10.1111/cns.14230] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2022] [Revised: 03/16/2023] [Accepted: 04/10/2023] [Indexed: 04/28/2023] Open
Abstract
AIMS We aimed to identify the neurotrophic activities of apigenin (4',5,7-trihydroxyflavone) via its coordination with brain-derived neurotrophic factor (BNDF) and an elevated signaling of tyrosine kinase receptor B (Trk B receptor). METHODS The direct binding of apigenin to BDNF was validated by ultrafiltration and biacore assay. Neurogenesis, triggered by apigenin and/or BDNF, was determined in cultured SH-SY5Y cells and rat cortical neurons. The amyloid-beta (Aβ)25-35 -induced cellular stress was revealed by propidium iodide staining, mitochondrial membrane potential, bioenergetic analysis, and formation of reactive oxygen species levels. Activation of Trk B signaling was tested by western blotting. RESULTS Apigenin and BDNF synergistically maintained the cell viability and promoted neurite outgrowth of cultured neurons. In addition, the BDNF-induced neurogenesis of cultured neurons was markedly potentiated by applied apigenin, including the induced expressions of neurofilaments, PSD-95 and synaptotagmin. Moreover, the synergy of apigenin and BDNF alleviated the (Aβ)25-35 -induced cytotoxicity and mitochondrial dysfunction. The synergy could be accounted by phosphorylation of Trk B receptor, and which was fully blocked by a Trk inhibitor K252a. CONCLUSION Apigenin potentiates the neurotrophic activities of BDNF through direct binding, which may serve as a possible treatment for its curative efficiency in neurodegenerative diseases and depression.
Collapse
Affiliation(s)
- Alex Xiong Gao
- Shenzhen Key Laboratory of Edible and Medicinal BioresourcesHKUST Shenzhen Research InstituteShenzhenChina
- Division of Life Science, Center for Chinese Medicine and State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyHong KongChina
| | - Tracy Chen‐Xi Xia
- Shenzhen Key Laboratory of Edible and Medicinal BioresourcesHKUST Shenzhen Research InstituteShenzhenChina
- Division of Life Science, Center for Chinese Medicine and State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyHong KongChina
| | - Lish Sheng‐Ying Lin
- Shenzhen Key Laboratory of Edible and Medicinal BioresourcesHKUST Shenzhen Research InstituteShenzhenChina
- Division of Life Science, Center for Chinese Medicine and State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyHong KongChina
| | - Tina Ting‐Xia Dong
- Shenzhen Key Laboratory of Edible and Medicinal BioresourcesHKUST Shenzhen Research InstituteShenzhenChina
- Division of Life Science, Center for Chinese Medicine and State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyHong KongChina
| | - Karl Wah‐Keung Tsim
- Shenzhen Key Laboratory of Edible and Medicinal BioresourcesHKUST Shenzhen Research InstituteShenzhenChina
- Division of Life Science, Center for Chinese Medicine and State Key Laboratory of Molecular NeuroscienceThe Hong Kong University of Science and TechnologyHong KongChina
| |
Collapse
|
3
|
Mießner H, Seidel J, Smith ESJ. In vitro models for investigating itch. Front Mol Neurosci 2022; 15:984126. [PMID: 36385768 PMCID: PMC9644192 DOI: 10.3389/fnmol.2022.984126] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2022] [Accepted: 10/10/2022] [Indexed: 12/04/2022] Open
Abstract
Itch (pruritus) is a sensation that drives a desire to scratch, a behavior observed in many animals. Although generally short-lasting and not causing harm, there are several pathological conditions where chronic itch is a hallmark symptom and in which prolonged scratching can induce damage. Finding medications to counteract the sensation of chronic itch has proven difficult due to the molecular complexity that involves a multitude of triggers, receptors and signaling pathways between skin, immune and nerve cells. While much has been learned about pruritus from in vivo animal models, they have limitations that corroborate the necessity for a transition to more human disease-like models. Also, reducing animal use should be encouraged in research. However, conducting human in vivo experiments can also be ethically challenging. Thus, there is a clear need for surrogate models to be used in pre-clinical investigation of the mechanisms of itch. Most in vitro models used for itch research focus on the use of known pruritogens. For this, sensory neurons and different types of skin and/or immune cells are stimulated in 2D or 3D co-culture, and factors such as neurotransmitter or cytokine release can be measured. There are however limitations of such simplistic in vitro models. For example, not all naturally occurring cell types are present and there is also no connection to the itch-sensing organ, the central nervous system (CNS). Nevertheless, in vitro models offer a chance to investigate otherwise inaccessible specific cell–cell interactions and molecular pathways. In recent years, stem cell-based approaches and human primary cells have emerged as viable alternatives to standard cell lines or animal tissue. As in vitro models have increased in their complexity, further opportunities for more elaborated means of investigating itch have been developed. In this review, we introduce the latest concepts of itch and discuss the advantages and limitations of current in vitro models, which provide valuable contributions to pruritus research and might help to meet the unmet clinical need for more refined anti-pruritic substances.
Collapse
Affiliation(s)
- Hendrik Mießner
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- Dermatological Skin Care, Beiersdorf AG, Hamburg, Germany
| | - Judith Seidel
- Dermatological Skin Care, Beiersdorf AG, Hamburg, Germany
| | - Ewan St. John Smith
- Department of Pharmacology, University of Cambridge, Cambridge, United Kingdom
- *Correspondence: Ewan St. John Smith,
| |
Collapse
|
4
|
Zhi WJ, Qiao SM, Zou Y, Peng RY, Yan HT, Ma LZ, Dong J, Zhao L, Yao BW, Zhao XL, Feng XX, Hu XJ, Wang LF. Low p-SYN1 (Ser-553) Expression Leads to Abnormal Neurotransmitter Release of GABA Induced by Up-Regulated Cdk5 after Microwave Exposure: Insights on Protection and Treatment of Microwave-Induced Cognitive Dysfunction. Curr Issues Mol Biol 2021; 44:206-221. [PMID: 35723394 PMCID: PMC8929049 DOI: 10.3390/cimb44010015] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/25/2021] [Revised: 12/22/2021] [Accepted: 12/27/2021] [Indexed: 11/23/2022] Open
Abstract
With the wide application of microwave technology, concerns about its health impact have arisen. The signal transmission mode of the central nervous system and neurons make it particularly sensitive to electromagnetic exposure. It has been reported that abnormal release of amino acid neurotransmitters is mediated by alteration of p-SYN1 after microwave exposure, which results in cognitive dysfunction. As the phosphorylation of SYN1 is regulated by different kinases, in this study we explored the regulatory mechanisms of SYN1 fluctuations following microwave exposure and its subsequent effect on GABA release, aiming to provide clues on the mechanism of cognitive impairment caused by microwave exposure. In vivo studies with Timm and H&E staining were adopted and the results showed abnormality in synapse formation and neuronal structure, explaining the previously-described deficiency in cognitive ability caused by microwave exposure. The observed alterations in SYN1 level, combined with the results of earlier studies, indicate that SYN1 and its phosphorylation status (ser-553 and ser62/67) may play a role in the abnormal release of neurotransmitters. Thus, the role of Cdk5, the upstream kinase regulating the formation of p-SYN1 (ser-553), as well as that of MEK, the regulator of p-SYN1 (ser-62/67), were investigated both in vivo and in vitro. The results showed that Cdk5 was a negative regulator of p-SYN1 (ser-553) and that its up-regulation caused a decrease in GABA release by reducing p-SYN1 (ser-553). While further exploration still needed to elaborate the role of p-SYN1 (ser-62/67) for neurotransmitter release, MEK inhibition had was no impact on p-Erk or p-SYN1 (ser-62/67) after microwave exposure. In conclusion, the decrease of p-SYN1 (ser-553) may result in abnormalities in vesicular anchoring and GABA release, which is caused by increased Cdk5 regulated through Calpain-p25 pathway after 30 mW/cm2 microwave exposure. This study provided a potential new strategy for the prevention and treatment of microwave-induced cognitive dysfunction.
Collapse
Affiliation(s)
- Wei-Jia Zhi
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China; (W.-J.Z.); (Y.Z.); (R.-Y.P.); (L.-Z.M.); (J.D.); (L.Z.); (B.-W.Y.); (X.-L.Z.)
| | - Si-Mo Qiao
- Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, China; (S.-M.Q.); (H.-T.Y.)
| | - Yong Zou
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China; (W.-J.Z.); (Y.Z.); (R.-Y.P.); (L.-Z.M.); (J.D.); (L.Z.); (B.-W.Y.); (X.-L.Z.)
| | - Rui-Yun Peng
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China; (W.-J.Z.); (Y.Z.); (R.-Y.P.); (L.-Z.M.); (J.D.); (L.Z.); (B.-W.Y.); (X.-L.Z.)
| | - Hai-Tao Yan
- Beijing Institute of Pharmacology and Toxicology, 27 Taiping Road, Beijing 100850, China; (S.-M.Q.); (H.-T.Y.)
| | - Li-Zhen Ma
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China; (W.-J.Z.); (Y.Z.); (R.-Y.P.); (L.-Z.M.); (J.D.); (L.Z.); (B.-W.Y.); (X.-L.Z.)
| | - Ji Dong
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China; (W.-J.Z.); (Y.Z.); (R.-Y.P.); (L.-Z.M.); (J.D.); (L.Z.); (B.-W.Y.); (X.-L.Z.)
| | - Li Zhao
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China; (W.-J.Z.); (Y.Z.); (R.-Y.P.); (L.-Z.M.); (J.D.); (L.Z.); (B.-W.Y.); (X.-L.Z.)
| | - Bin-Wei Yao
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China; (W.-J.Z.); (Y.Z.); (R.-Y.P.); (L.-Z.M.); (J.D.); (L.Z.); (B.-W.Y.); (X.-L.Z.)
| | - Xue-Long Zhao
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China; (W.-J.Z.); (Y.Z.); (R.-Y.P.); (L.-Z.M.); (J.D.); (L.Z.); (B.-W.Y.); (X.-L.Z.)
| | - Xin-Xing Feng
- Endocrine and Cardiovascular Center, Cardiovascular Institute and Fuwai Hospital of Chinese Academy of Medical Sciences, Beijing 100850, China;
| | - Xiang-Jun Hu
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China; (W.-J.Z.); (Y.Z.); (R.-Y.P.); (L.-Z.M.); (J.D.); (L.Z.); (B.-W.Y.); (X.-L.Z.)
- Correspondence: (X.-J.H.); (L.-F.W.)
| | - Li-Feng Wang
- Beijing Institute of Radiation Medicine, 27 Taiping Road, Beijing 100850, China; (W.-J.Z.); (Y.Z.); (R.-Y.P.); (L.-Z.M.); (J.D.); (L.Z.); (B.-W.Y.); (X.-L.Z.)
- Correspondence: (X.-J.H.); (L.-F.W.)
| |
Collapse
|
5
|
Tuftelin Is Required for NGF-Induced Differentiation of PC12 Cells. J Mol Neurosci 2019; 68:135-143. [PMID: 30903486 DOI: 10.1007/s12031-019-01292-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2018] [Accepted: 03/07/2019] [Indexed: 12/15/2022]
Abstract
Nerve growth factor (NGF) promotes pleiotropic gene transcription-dependent biological effects, in neuronal and non-neuronal cells, including survival, proliferation, differentiation, neuroprotection, pain, and angiogenesis. It is hypothesized that during odontogenesis, NGF may be implicated in morphogenetic and mineralization events by affecting proliferation and/or differentiation of dental cells. Tuftelin belongs to the enamel associated teeth proteins and is thought to play a role in enamel mineralization. We previously reported that tuftelin transcript and protein, which are ubiquitously expressed in various tissues of embryos, adults, and tumors, were significantly upregulated during NGF-induced PC12 differentiation. To further confirm the involvement of tuftelin in the differentiation process, we established a tuftelin-knockdown neuronal PC12 cell model, using a non-cytotoxic siRNA directed towards sequences at the 3' UTR of the tuftelin gene. Using real-time PCR, we quantified tuftelin mRNA expression and found that tuftelin siRNA, but not scrambled siRNA or transfection reagents, efficiently depleted about 60% of NGF-induced tuftelin mRNA transcripts. The effect of tuftelin siRNA was quantified up to 6 days of NGF-induced differentiation. Using immunofluorescence and western blot analyses, we also found a direct correlation between reduction of 60-80% in tuftelin protein expression and inhibition of about 50-70% in NGF-induced differentiation of the cells, as was detected after 3-6 days of treatment. These results demonstrate an important role for tuftelin in NGF-induced differentiation of PC12 cells. Tuftelin could be a useful target for drug development in disease where neurotrophin therapy is required.
Collapse
|
6
|
Growing Neural PC-12 Cell on Crosslinked Silica Aerogels Increases Neurite Extension in the Presence of an Electric Field. J Funct Biomater 2018; 9:jfb9020030. [PMID: 29677113 PMCID: PMC6023435 DOI: 10.3390/jfb9020030] [Citation(s) in RCA: 19] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2018] [Revised: 04/11/2018] [Accepted: 04/18/2018] [Indexed: 12/27/2022] Open
Abstract
Externally applied electrical stimulation (ES) has been shown to enhance the nerve regeneration process and to influence the directionality of neurite outgrowth. In addition, the physical and chemical properties of the substrate used for nerve-cell regeneration is critical in fostering regeneration. Previously, we have shown that polyurea-crosslinked silica aerogels (PCSA) exert a positive influence on the extension of neurites by PC-12 cells, a cell-line model widely used to study neurite extension and electrical excitability. In this work, we have examined how an externally applied electric field (EF) influences the extension of neurites in PC-12 cells grown on two substrates: collagen-coated dishes versus collagen-coated crosslinked silica aerogels. The externally applied direct current (DC) bias was applied in vitro using a custom-designed chamber containing polydimethysiloxane (PDMS) embedded copper electrodes to create an electric field across the substrate for the cultured PC-12 cells. Results suggest orientation preference towards the anode, and, on average, longer neurites in the presence of the applied DC bias than with 0 V DC bias. In addition, neurite length was increased in cells grown on silica-crosslinked aerogel when compared to cells grown on regular petri-dishes. These results further support the notion that PCSA is a promising material for nerve regeneration.
Collapse
|
7
|
Tatard VM, Venier-Julienne MC, Benoit JP, Menei P, Montero-Menei CN. In Vivo Evaluation of Pharmacologically Active Microcarriers Releasing Nerve Growth Factor and Conveying PC12 Cells. Cell Transplant 2017; 13:573-83. [PMID: 15565869 DOI: 10.3727/000000004783983675] [Citation(s) in RCA: 30] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Cell therapy will probably become a major therapeutic strategy in the coming years. Nevertheless, few cells survive transplantation when employed as a treatment for neuronal disorders. To address this problem, we have developed a new tool, the pharmacologically active microcarriers (PAM). PAM are biocompatible and biodegradable microparticles coated with cell adhesion molecules, conveying cells on their surface and presenting a controlled delivery of growth factor. Thus, the combined effect of growth factor and coating influences the transported cells by promoting their survival and differentiation and favoring their integration in the host tissue after their complete degradation. Furthermore, the released factor may also influence the microenvironment. In this study, we evaluated their efficacy using nerve growth factor (NGF)-releasing PAM and PC12 cells, in a Parkinson's disease paradigm. After implantation of NGF-releasing or unloaded PAM conveying PC12 cells, or PC12 cells alone, we studied cell survival, differentiation, and apoptosis, as well as behavior of the treated rats. We observed that the NGF-releasing PAM coated with two synthetic peptides (poly-D-lysine and fibronectin-like) induced PC12 cell differentiation and reduced cell death and proliferation. Moreover, the animals receiving this implant presented an improved amphetamine-induced rotational behavior. These findings indicate that PAM could be a promising strategy for cell therapy of neurological diseases and could be employed in other situations with fetal cell transplants or with stem cells.
Collapse
Affiliation(s)
- V M Tatard
- INSERM U 646, Laboratoire d'Ingénierie de la vectorisation particulaire, 10 rue André Boquel, 49100 Angers, France
| | | | | | | | | |
Collapse
|
8
|
Wang R, Yan F, Liao R, Wan P, Little PJ, Zheng W. Role of brain-derived neurotrophic factor and nerve growth factor in the regulation of Neuropeptide W in vitro and in vivo. Mol Cell Endocrinol 2017; 447:71-78. [PMID: 28249734 DOI: 10.1016/j.mce.2017.02.040] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/21/2016] [Revised: 02/03/2017] [Accepted: 02/25/2017] [Indexed: 01/24/2023]
Abstract
Nerve growth factor (NGF) and Brain-derived neurotrophic factor (BDNF) are neurotrophic factors involved in the growth, survival and functioning of neurons. In addition, a possible role of neurotrophins, particularly BDNF, in HPA axis hyperactivation has recently been proposed. Neuropeptide W (NPW) is an endogenous peptide ligand for the GPR7 and GPR8 and a stress mediator in the hypothalamus. It activates the HPA axis by working on hypothalamic corticotrophin-releasing hormone (CRH). No information is available about the interrelationships between neurotrophines like NGF/BDNF and NPW. We studied the effect and underlying mechanisms of NGF/BDNF on the production of NPW in PC12 cells and hypothalamus. NGF time- and concentration-dependently stimulated the expression of NPW in PC12 cells. The effect of NGF was blocked by the inhibition of PI3K/Akt signal pathway with specific inhibitors for PI3K or AktsiRNA for Akt while inhibition of ERK pathway had no effect. Moreover, BDNF concentration-dependently induced the expression of NPW mRNA and decreased the expression of NPY mRNA in primary cultured hypothalamic neurons which was also blocked by a PI3K kinase inhibitor. Finally, in vivo study showed that exogenous BDNF injected icv increased NPW production in the hypothalamus and this effect was reversed by a PI3 kinase inhibitor. These results and the fact that BDNF was able to stimulate the expression of CRH demonstrated that neurotrophines can modulate the expression of NPW in neuronal cells via the PI3K/Akt pathway and suggest that BDNF might be involved in functions of the HPA axis, at least in part by modulating the expression of NPW/NPY and CRH.
Collapse
Affiliation(s)
- Rikang Wang
- Faculty of Health Sciences, University of Macau, Macau, China; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center and School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China; National Pharmaceutical Engineering Center for Solid Preparation in Chinese Herbal Medicine, Jiangxi University of Traditional Chinese Medicine, Nanchang 330006, China; Shenzhen Key Laboratory for Anti-ageing and Regenerative Medicine, Health Science Center, Shenzhen University, Shenzhen 518060, China
| | - Fengxia Yan
- Faculty of Health Sciences, University of Macau, Macau, China; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center and School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Rifang Liao
- Faculty of Health Sciences, University of Macau, Macau, China; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center and School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China
| | - Pei Wan
- Faculty of Health Sciences, University of Macau, Macau, China
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, 20 Cornwall St, Woolloongabba, QLD, 4102, Australia
| | - Wenhua Zheng
- Faculty of Health Sciences, University of Macau, Macau, China; State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center and School of Pharmaceutical Sciences, Sun Yat-sen University, Guangzhou, China.
| |
Collapse
|
9
|
Crispo M, Dos Santos-Neto PC, Vilariño M, Mulet AP, de León A, Barbeito L, Menchaca A. RAPID COMMUNICATION: Nerve growth factor influences cleavage rate and embryo development in sheep. J Anim Sci 2017; 94:4447-4451. [PMID: 27898841 DOI: 10.2527/jas.2016-0736] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Recent information about Nerve growth factor (NGF), a protein traditionally associated to the nervous system that regulates survival and maturation of developing neurons, suggests that it may exert action also on different levels in the reproductive system. The aim of this study was to evaluate the effect of NGF added during in vitro oocyte maturation, fertilization or in vitro embryo development in sheep. Nerve growth factor was supplemented to the culture medium at 0, 100, or 1,000 ng/mL, during either in vitro maturation (Exp. 1), in vitro fertilization (Exp. 2), or in vitro culture (Exp. 3). In addition, NGF mRNA expression was determined in cumulus cells and oocytes. Nerve growth factor induced early cleavage when added during oocyte maturation or fertilization, improved embryo development when added during fertilization, and had no significant effect when added during embryo culture. In general, the effect was more evident with 100 rather than 1,000 ng/mL (P < 0.05). Expression of endogenous NGF was not detected in oocytes, and increased in cumulus cells when 1,000 ng/mL of NGF was added during fertilization, but not during maturation and embryo culture. In conclusion, the addition of NGF during oocyte maturation and fertilization affects in vitro cleavage and embryo development in sheep. We suggest a possible effect of this growth factor on oocyte maturation and mainly on the fertilization process.
Collapse
|
10
|
Back MJ, Lee HK, Lee JH, Fu Z, Son MW, Choi SZ, Go HS, Yoo S, Hwang SW, Kim DK. P2X1 Receptor-Mediated Ca 2+ Influx Triggered by DA-9801 Potentiates Nerve Growth Factor-Induced Neurite Outgrowth. ACS Chem Neurosci 2016; 7:1488-1498. [PMID: 27442785 DOI: 10.1021/acschemneuro.6b00082] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Nerve growth factor (NGF)-induced neuronal regeneration has emerged as a strategy to treat neuronal degeneration-associated disorders. However, direct NGF administration is limited by the occurrence of adverse effects at high doses of NGF. Therefore, development of a therapeutic strategy to promote the NGF trophic effect is required. In view of the lack of understanding of the mechanism for potentiating the NGF effect, this study investigated molecular targets of DA-9801, a well-standardized Dioscorea rhizome extract, which has a promoting effect on NGF. An increase in intracellular calcium ion level was induced by DA-9801, and chelation of extracellular calcium ions with ethylene-bis(oxyethylenenitrilo)tetraacetic acid (EGTA) suppressed the potentiating effect of DA-9801 on NGF-induced neurite outgrowth. In addition, EGTA treatment reduced the DA-9801-induced phosphorylation of extracellular signal-regulated kinase1/2 (ERK1/2), the major mediators of neurite outgrowth. To find which calcium ion-permeable channel contributes to the calcium ion influx induced by DA-9801, we treated PC12 cells with various inhibitors of calcium ion-permeable channels. NF449, a P2X1 receptor selective antagonist, significantly abolished the potentiating effect of DA-9801 on NGF-induced neurite outgrowth and abrogated the DA-9801-induced ERK1/2 phosphorylation. In addition, transfection with siRNA of P2X1 receptor significantly reduced the DA-9801-enhanced neurite outgrowth. In conclusion, calcium ion influx through P2X1 receptor mediated the promoting effect of DA-9801 on NGF-induced neurite outgrowth via ERK1/2 phosphorylation.
Collapse
Affiliation(s)
- Moon Jung Back
- Department
of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University; 84 Heukseok-ro, Dongjak-Ku, Seoul 06974, Republic of Korea
| | - Hae Kyung Lee
- Department
of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University; 84 Heukseok-ro, Dongjak-Ku, Seoul 06974, Republic of Korea
| | - Joo Hyun Lee
- Department
of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University; 84 Heukseok-ro, Dongjak-Ku, Seoul 06974, Republic of Korea
| | - Zhicheng Fu
- Department
of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University; 84 Heukseok-ro, Dongjak-Ku, Seoul 06974, Republic of Korea
| | - Mi Won Son
- Department of Research Planning & Management, Research Center of Dong-A ST Co., Ltd.; 21 Geumhwa-ro, 105 beon-gil, Giheung-gu, Yongin-si, Gyeonggi-do 446-905, Republic of Korea
| | - Sang Zin Choi
- Department of Research Planning & Management, Research Center of Dong-A ST Co., Ltd.; 21 Geumhwa-ro, 105 beon-gil, Giheung-gu, Yongin-si, Gyeonggi-do 446-905, Republic of Korea
| | - Hyo Sang Go
- Department of Research Planning & Management, Research Center of Dong-A ST Co., Ltd.; 21 Geumhwa-ro, 105 beon-gil, Giheung-gu, Yongin-si, Gyeonggi-do 446-905, Republic of Korea
| | - Sungjae Yoo
- Department
of Biomedical Sciences, Korea University College of Medicine, Seoul 136-705, Republic of Korea
| | - Sun Wook Hwang
- Department
of Biomedical Sciences, Korea University College of Medicine, Seoul 136-705, Republic of Korea
| | - Dae Kyong Kim
- Department
of Environmental and Health Chemistry, College of Pharmacy, Chung-Ang University; 84 Heukseok-ro, Dongjak-Ku, Seoul 06974, Republic of Korea
| |
Collapse
|
11
|
Zeng Z, Wang H, Shang F, Zhou L, Little PJ, Quirion R, Zheng W. Lithium ions attenuate serum-deprivation-induced apoptosis in PC12 cells through regulation of the Akt/FoxO1 signaling pathways. Psychopharmacology (Berl) 2016; 233:785-94. [PMID: 26626328 DOI: 10.1007/s00213-015-4168-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/26/2015] [Accepted: 11/10/2015] [Indexed: 11/26/2022]
Abstract
RATIONALE Lithium is currently used in the treatment of mental illness. We have previously reported that lithium stimulated the protein kinase B/Forkhead box O1 (Akt/FoxO1) pathway in rats. However, little information is available regarding its neuroprotective role of this pathway and underlying mechanisms. OBJECTIVES PC12 cells treated with serum deprivation were used as a toxicity model to study the protective effect of lithium and its underlying mechanisms. METHODS Cell viability was determined by methyl thiazolyl tetrazolium assay and Hoechst staining. FoxO1 subcellular location and its overexpression were used to study the underlying mechanisms. Various pathway inhibitors were used to investigate the possible pathways, while the phosphorylation of Akt and FoxO1 was analyzed by Western blot. RESULTS Lithium pretreatment dose-dependently reduced PC12 cell apoptosis induced by serum starvation. The protective effect of lithium was abolished by LY294002, a PI3K-specific inhibitor, and Akt inhibitor Akt inhibitor VIII, whereas mitogen-activated protein kinase kinase (MEK kinase) inhibitor U0126 had no effect. Lithium induced the phosphorylation of Akt and FoxO1 in a time- and concentration-dependent manner. Lithium-induced phosphorylation of Akt and FoxO1 is mediated by the PI3K/Akt pathway. Serum deprivation caused nuclear translocation of FoxO1 while application of lithium reversed the effect of serum deprivation. Moreover, overexpression of FoxO1 enhanced cell apoptosis induced by serum withdrawal. Finally, lithium was found to reduce the exogenous and endogenous FoxO1 protein levels in PC12 cells in a concentration-dependent fashion. CONCLUSIONS The protective effect of lithium against serum starvation cell death is mediated by the PI3K/Akt/FoxO1 pathway.
Collapse
Affiliation(s)
- Zhiwen Zeng
- Faculty of Health Science, University of Macau, Room 4021, Building E12, Avenida de Universidade Taipa, Macau, People's Republic of China
- Key Laboratory of Neurogenetics and Channelopathies of Guangdong Province and the Ministry of Education of China, Collaborative Innovation Center for Neurogenetics and Channelopathies, Institute of Neuroscience, The Second Affiliated Hospital of Guangzhou Medical University, Guangzhou, People's Republic of China
| | - Haitao Wang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center and Neurophamacology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510060, People's Republic of China
| | - Fu Shang
- State Key Laboratory of Ophthalmology, Zhongshan Ophthalmic Center and Neurophamacology, School of Pharmaceutical Sciences, Sun Yat-Sen University, Guangzhou, 510060, People's Republic of China
| | - Lihua Zhou
- Zhongshan Medical School, Sun Yat-Sen University, Guangzhou, 510060, People's Republic of China
| | - Peter J Little
- School of Pharmacy, Pharmacy Australia Centre of Excellence, The University of Queensland, 20 Cornwall St, Woolloongabba, QLD, 4102, Australia
| | - Remi Quirion
- Douglas Hospital Research Center, McGill University, Montreal, Canada
| | - Wenhua Zheng
- Faculty of Health Science, University of Macau, Room 4021, Building E12, Avenida de Universidade Taipa, Macau, People's Republic of China.
| |
Collapse
|
12
|
Sengupta T, Vinayagam J, Singh R, Jaisankar P, Mohanakumar KP. Plant-Derived Natural Products for Parkinson's Disease Therapy. ADVANCES IN NEUROBIOLOGY 2016; 12:415-96. [PMID: 27651267 DOI: 10.1007/978-3-319-28383-8_23] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Plant-derived natural products have made their own niche in the treatment of neurological diseases since time immemorial. Parkinson's disease (PD), the second most prevalent neurodegenerative disorder, has no cure and the treatment available currently is symptomatic. This chapter thoughtfully and objectively assesses the scientific basis that supports the increasing use of these plant-derived natural products for the treatment of this chronic and progressive disorder. Proper considerations are made on the chemical nature, sources, preclinical tests and their validity, and mechanisms of behavioural or biochemical recovery observed following treatment with various plants derived natural products relevant to PD therapy. The scientific basis underlying the neuroprotective effect of 6 Ayurvedic herbs/formulations, 12 Chinese medicinal herbs/formulations, 33 other plants, and 5 plant-derived molecules have been judiciously examined emphasizing behavioral, cellular, or biochemical aspects of neuroprotection observed in the cellular or animal models of the disease. The molecular mechanisms triggered by these natural products to promote cell survivability and to reduce the risk of cellular degeneration have also been brought to light in this study. The study helped to reveal certain limitations in the scenario: lack of preclinical studies in all cases barring two; heavy dependence on in vitro test systems; singular animal or cellular model to establish any therapeutic potential of drugs. This strongly warrants further studies so as to reproduce and confirm these reported effects. However, the current literature offers scientific credence to traditionally used plant-derived natural products for the treatment of PD.
Collapse
Affiliation(s)
- T Sengupta
- Division of Cell Biology & Physiology, Indian Institute of Chemical Biology (CSIR, Govt of India), 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700 032, India
| | - J Vinayagam
- Division of Chemistry, Indian Institute of Chemical Biology (CSIR, Govt of India), 4, Raja S.C. Mullick Road, Kolkata, 700 032, India
| | - R Singh
- Division of Cell Biology & Physiology, Indian Institute of Chemical Biology (CSIR, Govt of India), 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700 032, India
| | - P Jaisankar
- Division of Chemistry, Indian Institute of Chemical Biology (CSIR, Govt of India), 4, Raja S.C. Mullick Road, Kolkata, 700 032, India
| | - K P Mohanakumar
- Division of Cell Biology & Physiology, Indian Institute of Chemical Biology (CSIR, Govt of India), 4, Raja S.C. Mullick Road, Jadavpur, Kolkata, 700 032, India. .,Inter University Centre for Biomedical Research & Super Specialty Hospital, Mahatma Gandhi University Campus at Thalappady, Rubber Board PO, Kottayam, 686009, Kerala, India.
| |
Collapse
|
13
|
Vainshtein A, Veenman L, Shterenberg A, Singh S, Masarwa A, Dutta B, Island B, Tsoglin E, Levin E, Leschiner S, Maniv I, Pe’er L, Otradnov I, Zubedat S, Aga-Mizrachi S, Weizman A, Avital A, Marek I, Gavish M. Quinazoline-based tricyclic compounds that regulate programmed cell death, induce neuronal differentiation, and are curative in animal models for excitotoxicity and hereditary brain disease. Cell Death Discov 2015; 1:15027. [PMID: 27551459 PMCID: PMC4979516 DOI: 10.1038/cddiscovery.2015.27] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2015] [Accepted: 07/16/2015] [Indexed: 12/21/2022] Open
Abstract
Expanding on a quinazoline scaffold, we developed tricyclic compounds with biological activity. These compounds bind to the 18 kDa translocator protein (TSPO) and protect U118MG (glioblastoma cell line of glial origin) cells from glutamate-induced cell death. Fascinating, they can induce neuronal differentiation of PC12 cells (cell line of pheochromocytoma origin with neuronal characteristics) known to display neuronal characteristics, including outgrowth of neurites, tubulin expression, and NeuN (antigen known as 'neuronal nuclei', also known as Rbfox3) expression. As part of the neurodifferentiation process, they can amplify cell death induced by glutamate. Interestingly, the compound 2-phenylquinazolin-4-yl dimethylcarbamate (MGV-1) can induce expansive neurite sprouting on its own and also in synergy with nerve growth factor and with glutamate. Glycine is not required, indicating that N-methyl-D-aspartate receptors are not involved in this activity. These diverse effects on cells of glial origin and on cells with neuronal characteristics induced in culture by this one compound, MGV-1, as reported in this article, mimic the diverse events that take place during embryonic development of the brain (maintenance of glial integrity, differentiation of progenitor cells to mature neurons, and weeding out of non-differentiating progenitor cells). Such mechanisms are also important for protective, curative, and restorative processes that occur during and after brain injury and brain disease. Indeed, we found in a rat model of systemic kainic acid injection that MGV-1 can prevent seizures, counteract the process of ongoing brain damage, including edema, and restore behavior defects to normal patterns. Furthermore, in the R6-2 (transgenic mouse model for Huntington disease; Strain name: B6CBA-Tg(HDexon1)62Gpb/3J) transgenic mouse model for Huntington disease, derivatives of MGV-1 can increase lifespan by >20% and reduce incidence of abnormal movements. Also in vitro, these derivatives were more effective than MGV-1.
Collapse
Affiliation(s)
- A Vainshtein
- Department of Neuroscience, Technion – Israel Institute of Technology, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Haifa, Israel
| | - L Veenman
- Department of Neuroscience, Technion – Israel Institute of Technology, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Haifa, Israel
| | - A Shterenberg
- Technion – Israel Institute of Technology, Schulich Faculty of Chemistry, The Mallat Family Laboratory of Organic Chemistry, Haifa, Israel
| | - S Singh
- Technion – Israel Institute of Technology, Schulich Faculty of Chemistry, The Mallat Family Laboratory of Organic Chemistry, Haifa, Israel
| | - A Masarwa
- Technion – Israel Institute of Technology, Schulich Faculty of Chemistry, The Mallat Family Laboratory of Organic Chemistry, Haifa, Israel
| | - B Dutta
- Technion – Israel Institute of Technology, Schulich Faculty of Chemistry, The Mallat Family Laboratory of Organic Chemistry, Haifa, Israel
| | - B Island
- Technion – Israel Institute of Technology, Schulich Faculty of Chemistry, The Mallat Family Laboratory of Organic Chemistry, Haifa, Israel
| | - E Tsoglin
- Technion – Israel Institute of Technology, Schulich Faculty of Chemistry, The Mallat Family Laboratory of Organic Chemistry, Haifa, Israel
| | - E Levin
- Department of Neuroscience, Technion – Israel Institute of Technology, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Haifa, Israel
| | - S Leschiner
- Department of Neuroscience, Technion – Israel Institute of Technology, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Haifa, Israel
| | - I Maniv
- Department of Neuroscience, Technion – Israel Institute of Technology, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Haifa, Israel
| | - L Pe’er
- Department of Neuroscience, Technion – Israel Institute of Technology, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Haifa, Israel
| | - I Otradnov
- Department of Neuroscience, Technion – Israel Institute of Technology, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Haifa, Israel
| | - S Zubedat
- Department of Physiology, Technion – Israel Institute of Technology, The Behavioral Neuroscience Laboratory, Faculty of Medicine and Emek Medical Center, Haifa, Israel
| | - S Aga-Mizrachi
- Department of Physiology, Technion – Israel Institute of Technology, The Behavioral Neuroscience Laboratory, Faculty of Medicine and Emek Medical Center, Haifa, Israel
| | - A Weizman
- Tel Aviv University, Sackler Faculty of Medicine, The Felsenstein Medical Research Center, Geha Mental Health Center, Tel Aviv, Israel
| | - A Avital
- Department of Physiology, Technion – Israel Institute of Technology, The Behavioral Neuroscience Laboratory, Faculty of Medicine and Emek Medical Center, Haifa, Israel
| | - I Marek
- Technion – Israel Institute of Technology, Schulich Faculty of Chemistry, The Mallat Family Laboratory of Organic Chemistry, Haifa, Israel
| | - M Gavish
- Department of Neuroscience, Technion – Israel Institute of Technology, Faculty of Medicine, Rappaport Family Institute for Research in the Medical Sciences, Haifa, Israel
| |
Collapse
|
14
|
Jang Y, Lee MH, Lee J, Jung J, Lee SH, Yang DJ, Kim BW, Son H, Lee B, Chang S, Mori Y, Oh U. TRPM2 mediates the lysophosphatidic acid-induced neurite retraction in the developing brain. Pflugers Arch 2014; 466:1987-98. [PMID: 24413888 DOI: 10.1007/s00424-013-1436-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2013] [Revised: 12/23/2013] [Accepted: 12/23/2013] [Indexed: 01/12/2023]
Abstract
Intracellular Ca(2+) signal is a key regulator of axonal growth during brain development. As transient receptor potential (TRP) channels are permeable to Ca(2+) and mediate numerous brain functions, it is conceivable that many TRP channels would regulate neuronal differentiation. We therefore screened TRP channels that are involved in the regulation of neurite growth. Among the TRP channels, the Trpm2 level was inversely associated with neurite growth. TRPM2 was highly expressed in embryonic brain. Pharmacological perturbation or knockdown of TRPM2 markedly increased the axonal growth, whereas its overexpression inhibited the axonal growth. Addition of ADP ribose, an endogenous activator of TRPM2, to PC12 cells significantly repressed the axonal growth. TRPM2 was actively involved in the neuronal retraction induced by cerebrospinal fluid-rich lysophosphatidic acid (LPA). More importantly, neurons isolated from the brain of Trpm2-deficient mice have significantly longer neurites with a greater number of spines than those obtained from the brain of wild-type mice. Therefore, we conclude that TRPM2 mediates the LPA-induced suppression of axonal growth, which provides a long-sought mechanism underlying the effect of LPA on neuronal development.
Collapse
Affiliation(s)
- Yongwoo Jang
- Channel Research Center, CRI, College of Pharmacy, Seoul National University, 1 Gwanak-ro, Gwanak-gu, Seoul, 151-742, Republic of Korea
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
15
|
Nardicchi V, Ferrini M, Pilolli F, Angeli EB, Persichetti E, Beccari T, Mannucci R, Arcuri C, Donato R, Dorman RV, Goracci G. NGF Induces the Expression of Group IIA Secretory Phospholipase A2 in PC12 Cells: The Newly Synthesized Enzyme Is Addressed to Growing Neurites. Mol Neurobiol 2014; 50:15-25. [DOI: 10.1007/s12035-013-8621-5] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2013] [Accepted: 12/15/2013] [Indexed: 12/31/2022]
|
16
|
Antibodies directed to Neisseria gonorrhoeae impair nerve growth factor-dependent neurite outgrowth in Rat PC12 cells. J Mol Neurosci 2013; 52:353-65. [PMID: 24203572 DOI: 10.1007/s12031-013-0156-8] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2013] [Accepted: 10/17/2013] [Indexed: 12/17/2022]
Abstract
In children born from mothers with prenatal infections with the Gram-negative bacterium Neisseria gonorrhoeae, schizophrenia risk is increased in later life. Since cortical neuropil formation is frequently impaired during this disease, actions of a rabbit polyclonal antiserum directed to N. gonorrhoeae on neurite outgrowth in nerve growth factor-stimulated PC12 cells were investigated here. It turned out that 10 μg/ml of the antiserum leads indeed to a significant reduction in neurite outgrowth, whereas an antiserum directed to Neisseria meningitidis had no such effect. Furthermore, reduction in neurite outgrowth could be reversed by the neuroleptic drugs haloperidol, clozapine, risperidone, and olanzapine. On the molecular level, the observed effects seem to include the known neuritogenic transcription factors FoxO3a and Stat3, since reduced neurite outgrowth caused by the antiserum was accompanied by a reduced phosphorylation of both factors. In contrast, restitution of neurite outgrowth by neuroleptic drugs revealed no correlation to the phosphorylation state of these factors. The present report gives a first hint that bacterial infections could indeed lead to impaired neuropil formation in vitro; however, the in vivo relevance of this finding for schizophrenia pathogenesis remains to be clarified in the future.
Collapse
|
17
|
Nagahara Y, Suzuki E, Sekine Y, Uchiro H, Yoshimi Y, Shinomiya T, Ikekita M. SUTAF, a novel β-methoxyacrylate derivative, promotes neurite outgrowth with extracellular signal-regulated kinase and c-jun N-terminal kinase activation. Eur J Pharmacol 2012; 694:53-9. [PMID: 22975290 DOI: 10.1016/j.ejphar.2012.08.018] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/12/2012] [Revised: 08/24/2012] [Accepted: 08/27/2012] [Indexed: 10/27/2022]
Abstract
β-Methoxyacrylate antibiotics are well known to inhibit the fungal and yeast mitochondrial respiratory chain. In addition, β-methoxyacrylates are reported to suppress the proliferation of mammalian cancer cells. Differentiation and cell-cycle arrest are closely related. The cell cycle of proliferating cells is suppressed before differentiation. In this study, we synthesized a β-methoxyacrylate analog and treated neuronal differential model cells with it. We then estimated β-methoxyacrylate's neurotrophic effect by inhibiting cell proliferation so as to orient neuronal differentiation. SUTAF-027-a novel β-methoxyacrylate derivative, arrested the cell cycle and thereby suppressed the proliferation of PC12 rat pheochromocytoma cells and mouse neuroblastoma Neuro2a cells at very low treatment doses, as low as 1nM. However, a single SUTAF-027 treatment did not affect neuritogenesis. Surprisingly, however, co-treatment of SUTAF-027 and nerve growth factor (NGF) significantly augmented the NGF-induced neurite outgrowth of PC12. On the other hand, a single treatment of 1nM SUTAF-027 induced neurite outgrowth in Neuro2a cells. Further signal transduction mechanism studies revealed that SUTAF-027 induced the phosphorylation of extracellular signal-regulated kinase (ERK) and slight phosphorylation of c-jun N-terminal kinase (JNK). Moreover, inhibition of ERK and JNK blocked SUTAF-027-augmented neurite outgrowth. These results suggested that the novel β-methoxyacrylate analog SUTAF-027 augmented neurite outgrowth by arresting the cell cycle and activating the ERK and JNK pathways.
Collapse
Affiliation(s)
- Yukitoshi Nagahara
- Department of Biotechnology, College of Science and Engineering, Tokyo Denki University, Hatoyama, Hiki-gun, Saitama 350-0394, Japan.
| | | | | | | | | | | | | |
Collapse
|
18
|
Sirabella R, Secondo A, Pannaccione A, Molinaro P, Formisano L, Guida N, Di Renzo G, Annunziato L, Cataldi M. ERK1/2, p38, and JNK regulate the expression and the activity of the three isoforms of the Na+ /Ca2+ exchanger, NCX1, NCX2, and NCX3, in neuronal PC12 cells. J Neurochem 2012; 122:911-22. [PMID: 22708976 DOI: 10.1111/j.1471-4159.2012.07838.x] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/01/2022]
Abstract
We evaluated whether changes in expression and activity of the three sodium/calcium exchanger isoforms, NCX1, NCX2, and NCX3 occurred in PC12 cells when the extracellular-signal-regulated kinases 1/2 (ERK1/2), c-Jun N-terminal kinases (JNK), and p38 mitogen-activated protein kinases (MAPKs) were silenced, pharmacologically blocked, or activated with nerve growth factor (NGF). Several findings suggesting that MAPKs control NCX emerged: (1) A decrease in NCX1 and NCX3 basal expression occurred when JNK or MEK1, the extracellular-signal-regulated kinases 1/2 upstream activator, were pharmacologically blocked, respectively; (2) NGF increased cAMP response element-binding 1 (CREB1) and Specificity Protein 1 (Sp1) binding to ncx1 promoter and CREB1 binding to two different sequences close to ncx2 transcription start site on genomic DNA; (3) An up-regulation of NCX1 and NCX3, abrogated upon either MEK1 or p38 blockade, and a down-regulation of NCX2, abolished upon p38 blockade, occurred upon NGF-induced MAPK activation. The NCX1 up-regulation was abolished upon either CREB1 or Sp1 silencing, whereas NCX2 down-regulation was abrogated only by CREB1 silencing. The NCX3 up-regulation was unaffected by CREB1 or Sp1 silencing and abolished upon proteasomal inhibition; (4) Whole-cell Na(+) /Ca(2+) exchange decreased when MEK1 and JNK were blocked and increased when MAPKs were activated by NGF. Collectively, these results demonstrate a MAPK-dependent regulation of NCX expression and activity which could be relevant in mediating some of the effects of MAPKs in neurons.
Collapse
|
19
|
Neurotrophic effects of a cyanine dye via the PI3K-Akt pathway: attenuation of motor discoordination and neurodegeneration in an ataxic animal model. PLoS One 2011; 6:e17137. [PMID: 21347252 PMCID: PMC3037960 DOI: 10.1371/journal.pone.0017137] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/15/2010] [Accepted: 01/21/2011] [Indexed: 01/25/2023] Open
Abstract
BACKGROUND Neurotrophic factors may be future therapeutic agents for neurodegenerative disease. In the screening of biologically active molecules for neurotrophic potency, we found that a photosensitizing cyanine dye, NK-4, had remarkable neurotrophic activities and was a potent radical scavenger. METHODOLOGY/PRINCIPAL FINDINGS In this study, we evaluated the effect of NK-4 on the protection of neurons against oxidative damage and investigated the associated intracellular signaling pathways. Subsequently, we evaluated the effect of NK-4 in an animal model of neurodegeneration. In vitro, NK-4 showed dose-dependent protection of PC12 cells from toxicity induced by oxidative stress caused by hydrogen peroxide (H(2)O(2)) or 6-hydroxydopamine (6-OHDA). Comparison of extracellular signal-regulated kinase signaling pathways between treatment with NK-4 and nerve growth factor (NGF) using K252a, an inhibitor of the NGF receptor TrkA, revealed that NK-4 activity occurs independently of NGF receptors. LY294002, a phosphatidylinositol 3-kinase (PI3K) inhibitor, blocked the protective effect of NK-4, and NK-4 caused activation of Akt/protein kinase B, a downstream effector of PI3K. These results suggest that the neuroprotective effects of NK-4 are mediated by the PI3K-Akt signaling pathway. NK-4 treatment also attenuated stress-induced activation of SAPK/JNK, which suggests that NK-4 activates a survival signaling pathway and inhibits stress-activated apoptotic pathways independently of the TrkA receptor in neuronal cells. In vivo, administration of NK-4 improved motor coordination in genetic ataxic hamsters, as assessed by rota-rod testing. Histological analysis showed that cerebellar atrophy was significantly attenuated by NK-4 treatment. Notably, the Purkinje cell count in the treated group was threefold higher than that in the vehicle group. CONCLUSIONS/SIGNIFICANCE These results suggest that NK-4 is a potential agent for therapy for neurodegenerative disorders based on the activation of survival signaling pathways.
Collapse
|
20
|
Ferrini M, Nardicchi V, Mannucci R, Arcuri C, Nicoletti I, Donato R, Goracci G. Effect of NGF on the subcellular localization of group IIA secretory phospholipase A(2) (GIIA) in PC12 cells: role in neuritogenesis. Neurochem Res 2010; 35:2168-74. [PMID: 21125328 DOI: 10.1007/s11064-010-0345-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/08/2010] [Indexed: 01/01/2023]
Abstract
Phospholipases A(2) (PLA(2)s) are involved in neuritogenesis but the identity of the isoforms(s) contributing to this process is still not defined. Several reports have focused on secretory PLA(2)s (sPLA(2)) as the administration of exogenous sPLA(2)s to PC12 neuronal cells stimulates neurite outgrowth. The present study demonstrates that the endogenous group IIA sPLA(2) (GIIA), constitutively expressed in mammalian neural cells, changes its subcellular localization when PC12 cells are induced to differentiate by NGF treatment. Indeed, confocal analysis showed a time-dependent accumulation of GIIA in growth cones and neurite tips. Under identical conditions the subcellular distribution of another isoform (GV) was unaffected by NGF. Contrary to GX, another sPLA(2) isoform expressed by PC12 cells, the contribution of GIIA to neuritogenesis does not require its release in the extracellular medium.
Collapse
Affiliation(s)
- M Ferrini
- Departments of Experimental Medicine and Biochemical Sciences, University of Perugia, Via del Giochetto, 06126 Perugia, Italy
| | | | | | | | | | | | | |
Collapse
|
21
|
Yeyeodu S, Gilyazova N, Huh EY, Dandepally SR, Oldham C, Williams A, Ibeanu G. A trifluoromethyl analog of verbenachalcone promotes neurite outgrowth and cell proliferation of NeuroScreen-1 cells. Cell Mol Neurobiol 2010; 31:145-53. [PMID: 20848307 DOI: 10.1007/s10571-010-9563-3] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/14/2010] [Accepted: 08/27/2010] [Indexed: 12/12/2022]
Abstract
Past research has shown that natural products of plant and marine origins and their congeners enhance the actions of neuritogenic factors of the central nervous system (CNS) such as nerve growth factor (NGF). However, the role of fluorine substitutions in their structure-activity relationship (SAR) has not been explored. We have synthesized a trifluoromethyl analog of verbenachalcone (VC), a pharmacologically active natural compound previously shown to potentiate NGF activity. This analog, designated C278, enhances neurite outgrowth and proliferation of NeuroScreen-1™ (NS-1) cells, a subclone of PC12 pheochromocytoma cells. C278 increases the percentage of neurite bearing cells in the presence of suboptimal doses of NGF in comparison with controls treated with NGF alone. In addition, C278 stimulates cell growth in reduced serum and serum-free cell culture conditions based on our observation of increases in cell number and metabolic assessment with MTT reduction and resazurin assays. The addition of C278 partially restored inhibition of NGF-induced neurite outgrowth by the mitogen-activated protein kinase kinase (MEK) inhibitors PD98059 and U0126. Short-term sequential exposure of cells to U0126, C278, and NGF enhanced phosphorylation of extracellular signal-regulated kinase (ERK) in comparison with cells treated with only the MEK inhibitor and NGF. C278 also attenuated cell growth arrest caused by exposure to PD98059, U0126 and the phosphatidylinositol-3 kinase (PI3K) inhibitor, LY294002 but did not alter phosphorylation of Akt, a classic downstream target of PI3K during cell survival. These data suggest that C278 promotes NGF-dependent neurite outgrowth in NS-1 cells through a MEK signaling pathway by a mechanism that alters short-term activation of ERK. In contrast, C278 promotes PI3K-mediated survival independently of Akt phosphorylation.
Collapse
Affiliation(s)
- Susan Yeyeodu
- Biomanufacturing Research Institute and Technology Enterprise, Durham, NC, USA
| | | | | | | | | | | | | |
Collapse
|
22
|
Chen MC, Lin H, Hsu FN, Huang PH, Lee GS, Wang PS. Involvement of cAMP in nerve growth factor-triggered p35/Cdk5 activation and differentiation in PC12 cells. Am J Physiol Cell Physiol 2010; 299:C516-27. [PMID: 20463173 DOI: 10.1152/ajpcell.00534.2009] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The signaling mechanisms underlying cell differentiation have been extensively studied with the use of rat PC12 cells as a model system. Nerve growth factor (NGF) is a trophic factor inducing PC12 cell differentiation through the activation of the p35/cyclin-dependent kinase 5 (Cdk5) complex. It has been reported that adenylyl cyclase activation and cAMP production may be involved in NGF-dependent actions. Our previous results indicate that cAMP activates the p35/Cdk5 complex in reproductive cells. Therefore, the role of cAMP in NGF-triggered p35/Cdk5 activation and PC12 differentiation was interesting to explore. Our results indicate that roscovitine, a molecular inhibitor of Cdk5, blocks cAMP-triggered PC12 differentiation, which was evaluated by neurite initiation, a decrease in proliferation, and cell cycle G(1) arrest. The following data show that cAMP treatment increased Cdk5 activity through p35 upregulation. cAMP downstream components, protein kinase A (PKA) and phosphorylated cAMP response element binding protein (CREB), are involved in this regulation. The immunocytochemical results indicate that PKA inhibition disrupted cAMP-triggered p35/Cdk5 localization in PC12 cells. In addition, adenylyl cyclase inhibition was found to diminish NGF-induced intracellular cAMP production, CREB phosphorylation, and p35 expression. The cAMP antagonist and the PKA inhibitors reduced NGF-induced p35 expression. Finally, NGF-triggered PC12 differentiation was partially decreased by adenylyl cyclase or PKA inhibitors. In conclusion, these results demonstrate that cAMP may play a role in NGF-p35/Cdk5-dependent PC12 differentiation.
Collapse
Affiliation(s)
- Mei-Chih Chen
- Dept. of Physiology, School of Medicine, National Yang Ming University, Taipei 11221, Taiwan, Republic of China
| | | | | | | | | | | |
Collapse
|
23
|
Normalization with genes encoding ribosomal proteins but not GAPDH provides an accurate quantification of gene expressions in neuronal differentiation of PC12 cells. BMC Genomics 2010; 11:75. [PMID: 20113474 PMCID: PMC2831847 DOI: 10.1186/1471-2164-11-75] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2009] [Accepted: 01/29/2010] [Indexed: 12/14/2022] Open
Abstract
Background Gene regulation at transcript level can provide a good indication of the complex signaling mechanisms underlying physiological and pathological processes. Transcriptomic methods such as microarray and quantitative real-time PCR require stable reference genes for accurate normalization of gene expression. Some but not all studies have shown that housekeeping genes (HGKs), β-actin (ACTB) and glyceraldehyde-3-phosphate dehydrogenase (GAPDH), which are routinely used for normalization, may vary significantly depending on the cell/tissue type and experimental conditions. It is currently unclear if these genes are stably expressed in cells undergoing drastic morphological changes during neuronal differentiation. Recent meta-analysis of microarray datasets showed that some but not all of the ribosomal protein genes are stably expressed. To test the hypothesis that some ribosomal protein genes can serve as reference genes for neuronal differentiation, a genome-wide analysis was performed and putative reference genes were identified based on stability of expressions. The stabilities of these potential reference genes were then analyzed by reverse transcription quantitative real-time PCR in six differentiation conditions. Results Twenty stably expressed genes, including thirteen ribosomal protein genes, were selected from microarray analysis of the gene expression profiles of GDNF and NGF induced differentiation of PC12 cells. The expression levels of these candidate genes as well as ACTB and GAPDH were further analyzed by reverse transcription quantitative real-time PCR in PC12 cells differentiated with a variety of stimuli including NGF, GDNF, Forskolin, KCl and ROCK inhibitor, Y27632. The performances of these candidate genes as stable reference genes were evaluated with two independent statistical approaches, geNorm and NormFinder. Conclusions The ribosomal protein genes, RPL19 and RPL29, were identified as suitable reference genes during neuronal differentiation of PC12 cells, regardless of the type of differentiation conditions. The combination of these two novel reference genes, but not the commonly used HKG, GAPDH, allows robust and accurate normalization of differentially expressed genes during PC12 differentiation.
Collapse
|
24
|
Yue J, Wei W, Lam CMC, Zhao YJ, Dong M, Zhang LR, Zhang LH, Lee HC. CD38/cADPR/Ca2+ pathway promotes cell proliferation and delays nerve growth factor-induced differentiation in PC12 cells. J Biol Chem 2009; 284:29335-42. [PMID: 19696022 DOI: 10.1074/jbc.m109.049767] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022] Open
Abstract
Intracellular Ca(2+) mobilization plays an important role in a wide variety of cellular processes, and multiple second messengers are responsible for mediating intracellular Ca(2+) changes. Here we explored the role of one endogenous Ca(2+)-mobilizing nucleotide, cyclic adenosine diphosphoribose (cADPR), in the proliferation and differentiation of neurosecretory PC12 cells. We found that cADPR induced Ca(2+) release in PC12 cells and that CD38 is the main ADP-ribosyl cyclase responsible for the acetylcholine (ACh)-induced cADPR production in PC12 cells. In addition, the CD38/cADPR signaling pathway is shown to be required for the ACh-induced Ca(2+) increase and cell proliferation. Inhibition of the pathway, on the other hand, accelerated nerve growth factor (NGF)-induced neuronal differentiation in PC12 cells. Conversely, overexpression of CD38 increased cell proliferation but delayed NGF-induced differentiation. Our data indicate that cADPR plays a dichotomic role in regulating proliferation and neuronal differentiation of PC12 cells.
Collapse
Affiliation(s)
- Jianbo Yue
- Department of Physiology, The University of Hong Kong, Hong Kong.
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Xie J, Chen L, Varadan VK, Yancey J, Srivatsan M. The effects of functional magnetic nanotubes with incorporated nerve growth factor in neuronal differentiation of PC12 cells. NANOTECHNOLOGY 2008; 19:105101. [PMID: 21817692 DOI: 10.1088/0957-4484/19/10/105101] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/31/2023]
Abstract
In this in vitro study the efficiency of magnetic nanotubes to bind with nerve growth factor (NGF) and the ability of NGF-incorporated magnetic nanotubes to release the bound NGF are investigated using rat pheochromocytoma cells (PC12 cells). It is found that functional magnetic nanotubes with NGF incorporation enabled the differentiation of PC12 cells into neurons exhibiting growth cones and neurite outgrowth. Microscope observations show that filopodia extending from neuron growth cones were in close proximity to the NGF-incorporated magnetic nanotubes, at times appearing to extend towards or into them. These results show that magnetic nanotubes can be used as a delivery vehicle for NGF and thus may be exploited in attempts to treat neurodegenerative disorders such as Parkinson's disease with neurotrophins. Further neurite outgrowth can be controlled by manipulating magnetic nanotubes with external magnetic fields, thus helping in directed regeneration.
Collapse
Affiliation(s)
- Jining Xie
- Nanomaterials and Nanotubes Research Laboratory, College of Engineering, University of Arkansas, Fayetteville, AR 72701, USA
| | | | | | | | | |
Collapse
|
26
|
Spinsanti P, De Vita T, Caruso A, Melchiorri D, Misasi R, Caricasole A, Nicoletti F. Differential activation of the calcium/protein kinase C and the canonical beta-catenin pathway by Wnt1 and Wnt7a produces opposite effects on cell proliferation in PC12 cells. J Neurochem 2007; 104:1588-98. [PMID: 17988238 DOI: 10.1111/j.1471-4159.2007.05111.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023]
Abstract
We examined the effect of Wnt1 and Wnt7a on cell proliferation using undifferentiated PC12 cells, which originate from the neural crest and are widely employed as a neuronal cell model. Heterologous expression of Wnt1 enhanced [3H]thymidine incorporation and expression of cyclin D1 and cylin E in PC12 cells. Opposite effects were observed in PC12 cells expressing Wnt7a. Searching for the mechanisms underlying the opposite effects of Wnt1 and Wnt7a on PC12 cell proliferation, we examined the activation of the canonical beta-catenin/T-cell-lymphoid enhancer-binding protein transcription factor pathway and the 'calcium pathway' by co-transfecting the cells with a reporter gene controlled by either T-cell-lymphoid enhancer-binding protein transcription factor or the calcium-activated transcription factor, NFAT. Wnt1 and Wnt7a activated both pathways, but to a different extent. While Wnt1 preferentially activated the calcium pathway, Wnt7a mainly activated the canonical pathway. Pharmacological inhibition of protein kinase C, which is a component of the calcium pathway, abrogated the increase in cell proliferation induced by Wnt1 without affecting the antiproliferative action of Wnt7a. The action of Wnt7a was instead occluded by lithium ions, which mimic the activation of the canonical pathway, and was largely reduced by Dickkopf-1, which acts as an inhibitor of the canonical pathway. In addition, expression of a constitutively active mutant of beta-catenin potently activated the canonical Wnt pathway and reduced [3H]thymidine incorporation. These data challenge the view that the canonical Wnt pathway invariably supports cell growth and suggest that, at least in PC12 cells, cell proliferation is regulated by the balance between the calcium/protein kinase C pathway and the canonical pathway.
Collapse
Affiliation(s)
- Paola Spinsanti
- Department of Human Physiology and Pharmacology, University of Rome La Sapienza, Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
27
|
Gambino F, Pavlowsky A, Béglé A, Dupont JL, Bahi N, Courjaret R, Gardette R, Hadjkacem H, Skala H, Poulain B, Chelly J, Vitale N, Humeau Y. IL1-receptor accessory protein-like 1 (IL1RAPL1), a protein involved in cognitive functions, regulates N-type Ca2+-channel and neurite elongation. Proc Natl Acad Sci U S A 2007; 104:9063-8. [PMID: 17502602 PMCID: PMC1885628 DOI: 10.1073/pnas.0701133104] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/07/2007] [Indexed: 11/18/2022] Open
Abstract
Null mutations in the IL1-receptor accessory protein-like 1 gene (IL1RAPL1) are responsible for an inherited X-linked form of cognitive impairment. IL1RAPL1 protein physically interacts with neuronal calcium sensor-1 (NCS-1), but the functional impact of the IL1RAPL1/NCS-1 interaction remains unknown. Here, we demonstrate that stable expression of IL1RAPL1 in PC12 cells induces a specific silencing of N-type voltage-gated calcium channels (N-VGCC) activity that explains a secretion deficit observed in these IL1RAPL1 cells. Importantly, this modulation of VGCC activity is mediated by NCS-1. Indeed, a specific loss-of-function of N-VGCC was observed in PC12 cells overexpressing NCS-1, and a total recovery of N-VGCC activity was obtained by a down-regulation of NCS-1 in IL1RAPL1 cells. The functional relevance of the interaction between IL1RAPL1 and NCS-1 was also suggested by the reduction of neurite elongation observed in nerve growth factor (NGF)-treated IL1RAPL1 cells, a phenotype rescued by NCS-1 inactivation. Because both proteins are highly expressed in neurons, these results suggest that IL1RAPL1-related mental retardation could result from a disruption of N-VGCC and/or NCS-1-dependent synaptic and neuronal activities.
Collapse
Affiliation(s)
- Frédéric Gambino
- *Département Neurotransmission et Sécrétion Neuroendocrine, Institut des Neurosciences Cellulaires et Intégratives, Unité Mixte de Recherche 7168/LC2, Centre National de la Recherche Scientifique and Université Louis Pasteur, 5 Rue Blaise Pascal, 67084 Strasbourg, France
| | - Alice Pavlowsky
- Institut Cochin, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Université Paris Descartes, 75014 Paris, France
- Institut National de la Santé et de la Recherche Médicale, Unité 567, 75014 Paris, France; and
| | - Aurélie Béglé
- *Département Neurotransmission et Sécrétion Neuroendocrine, Institut des Neurosciences Cellulaires et Intégratives, Unité Mixte de Recherche 7168/LC2, Centre National de la Recherche Scientifique and Université Louis Pasteur, 5 Rue Blaise Pascal, 67084 Strasbourg, France
| | - Jean-Luc Dupont
- *Département Neurotransmission et Sécrétion Neuroendocrine, Institut des Neurosciences Cellulaires et Intégratives, Unité Mixte de Recherche 7168/LC2, Centre National de la Recherche Scientifique and Université Louis Pasteur, 5 Rue Blaise Pascal, 67084 Strasbourg, France
| | - Nadia Bahi
- Institut Cochin, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Université Paris Descartes, 75014 Paris, France
- Institut National de la Santé et de la Recherche Médicale, Unité 567, 75014 Paris, France; and
| | - Raphael Courjaret
- *Département Neurotransmission et Sécrétion Neuroendocrine, Institut des Neurosciences Cellulaires et Intégratives, Unité Mixte de Recherche 7168/LC2, Centre National de la Recherche Scientifique and Université Louis Pasteur, 5 Rue Blaise Pascal, 67084 Strasbourg, France
| | - Robert Gardette
- Institut National de la Santé et de la Recherche Médicale, Unité 549, IFR Broca Sainte Anne,2ter Rue d'Alesia, 75014 Paris, France
| | - Hassen Hadjkacem
- Institut Cochin, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Université Paris Descartes, 75014 Paris, France
- Institut National de la Santé et de la Recherche Médicale, Unité 567, 75014 Paris, France; and
| | - Henriette Skala
- Institut Cochin, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Université Paris Descartes, 75014 Paris, France
- Institut National de la Santé et de la Recherche Médicale, Unité 567, 75014 Paris, France; and
| | - Bernard Poulain
- *Département Neurotransmission et Sécrétion Neuroendocrine, Institut des Neurosciences Cellulaires et Intégratives, Unité Mixte de Recherche 7168/LC2, Centre National de la Recherche Scientifique and Université Louis Pasteur, 5 Rue Blaise Pascal, 67084 Strasbourg, France
| | - Jamel Chelly
- Institut Cochin, Centre National de la Recherche Scientifique, Unité Mixte de Recherche 8104, Université Paris Descartes, 75014 Paris, France
- Institut National de la Santé et de la Recherche Médicale, Unité 567, 75014 Paris, France; and
| | - Nicolas Vitale
- *Département Neurotransmission et Sécrétion Neuroendocrine, Institut des Neurosciences Cellulaires et Intégratives, Unité Mixte de Recherche 7168/LC2, Centre National de la Recherche Scientifique and Université Louis Pasteur, 5 Rue Blaise Pascal, 67084 Strasbourg, France
| | - Yann Humeau
- *Département Neurotransmission et Sécrétion Neuroendocrine, Institut des Neurosciences Cellulaires et Intégratives, Unité Mixte de Recherche 7168/LC2, Centre National de la Recherche Scientifique and Université Louis Pasteur, 5 Rue Blaise Pascal, 67084 Strasbourg, France
| |
Collapse
|
28
|
Vetrugno V, Cardinale A, Filesi I, Mattei S, Sy MS, Pocchiari M, Biocca S. KDEL-tagged anti-prion intrabodies impair PrP lysosomal degradation and inhibit scrapie infectivity. Biochem Biophys Res Commun 2005; 338:1791-7. [PMID: 16288721 DOI: 10.1016/j.bbrc.2005.10.146] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/19/2005] [Accepted: 10/23/2005] [Indexed: 11/29/2022]
Abstract
Transmissible spongiform encephalopathy or prion diseases are fatal neurodegenerative disorders characterized by the conversion of the cellular prion protein (PrPC) into the infectious scrapie isoform (PrPSc). We have recently demonstrated that anti-prion intrabodies targeted to the lumen of the endoplasmic reticulum provide a simple and effective means to inhibit the transport of PrPC to the cell surface. Here, we report that they completely block the traffic of mature full-length PrPC molecules, impair prion lysosomal degradation, and interfere with the early phase of scrapie formation. Since anti-prion intrabodies efficiently block PrPSc accumulation in vitro, we investigated whether they could also antagonize scrapie infectivity in vivo. We found that mice intracerebrally injected with KDEL-8H4-NGF-differentiated PC12 cells infected with scrapie neither develop scrapie clinical signs nor brain damage. Furthermore, no protease-resistant PrPSc is detectable in brains of inoculated animals. These results indicate that anti-prion intrabody strategy may be effective against prion infection.
Collapse
Affiliation(s)
- Vito Vetrugno
- Department of Neuroscience and Laboratory of Clinical Biochemistry, University of Tor Vergata, Via Montpellier 1, 00133 Rome, Italy
| | | | | | | | | | | | | |
Collapse
|
29
|
Chen YL, Law PY, Loh HH. Sustained activation of phosphatidylinositol 3-kinase/Akt/nuclear factor kappaB signaling mediates G protein-coupled delta-opioid receptor gene expression. J Biol Chem 2005; 281:3067-74. [PMID: 16316997 DOI: 10.1074/jbc.m506721200] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Expression of the delta-opioid receptor gene (dor) is tightly controlled during neuronal differentiation and developmental stages. Such distinct temporal and spatial expression of dor during development suggests a role for the delta-opioid receptor in early developmental events. However, little is known about intracellular signaling pathways that control dor expression. A well established cell line model for the study of gene expression during neuronal differentiation is the rat adrenal pheochromocytoma PC12 cell line. Here we found that the constitutively activated TrkA/phosphatidylinositol 3-kinase/Akt (protein kinase B)/NF-kappaB survival cascade mediates dor expression during nerve growth factor (NGF)-induced differentiation of PC12h cells. Biochemical experiments showed that constitutive phosphorylation of Akt and IkappaBalpha correlates with NGF-induced dor expression. Overexpression of the transcriptional activator NF-kappaB/p65 increased dor promoter activity. Overexpression of the NF-kappaB signaling super inhibitor mutant IkappaBalpha (S32A/S36A) abolished the effect of p65 and blocked NGF-induced activation of NF-kappaB signaling, resulting in a significant reduction in dor promoter activity. Treatment with SN50, an NF-kappaB-specific nuclear translocation peptide inhibitor, inhibited the translocation of NF-kappaB, resulting in a reduction of dor mRNA. The gel shift assay supported the fact that there exists an NF-kappaB-binding site on the dor promoter. RNA interference experiments using NF-kappaB/p65 small interfering RNA confirmed that NF-kappaB signaling is required for dor expression. Our findings not only provide a new mechanistic explanation for NGF-induced dor expression but also shed some light on the molecular mechanism of the temporal and spatial expression of dor and the roles of the delta-opioid receptor during neuronal differentiation.
Collapse
MESH Headings
- Active Transport, Cell Nucleus
- Animals
- Binding Sites
- Binding, Competitive
- Blotting, Western
- Cell Differentiation
- Cell Nucleus/metabolism
- Enzyme Activation
- Gene Expression Regulation, Enzymologic
- Genes, Reporter
- I-kappa B Proteins/metabolism
- Mutation
- NF-KappaB Inhibitor alpha
- NF-kappa B/metabolism
- Nerve Growth Factor/metabolism
- Neurons/metabolism
- Oligonucleotide Probes/chemistry
- PC12 Cells
- Peptides/pharmacology
- Phosphatidylinositol 3-Kinases/metabolism
- Phosphorylation
- Plasmids/metabolism
- Promoter Regions, Genetic
- Proto-Oncogene Proteins c-akt/metabolism
- RNA Interference
- RNA, Small Interfering/metabolism
- Rats
- Receptors, G-Protein-Coupled/biosynthesis
- Receptors, Opioid, delta/metabolism
- Reverse Transcriptase Polymerase Chain Reaction
- Signal Transduction
- Time Factors
- Transcription Factor RelA/metabolism
- Transcriptional Activation
- Transfection
Collapse
Affiliation(s)
- Yulong L Chen
- Department of Pharmacology, University of Minnesota Medical School, Minneapolis, Minnesota 55455, USA.
| | | | | |
Collapse
|
30
|
Bacsi A, Stanton GJ, Hughes TK, Kruze M, Boldogh I. Colostrinin-driven neurite outgrowth requires p53 activation in PC12 cells. Cell Mol Neurobiol 2005; 25:1123-39. [PMID: 16392041 DOI: 10.1007/s10571-005-8222-6] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/10/2004] [Accepted: 08/29/2005] [Indexed: 10/25/2022]
Abstract
1. Colostrinin (CLN) induces maturation and differentiation of murine thymocytes, promotes proliferation of peripheral blood leukocytes, induces immunomodulator cytokines, and ameliorates oxidative stress-mediated activation of c-Jun NH2-terminal kinases. 2. Here we report that upon treatment with CLN, medullary pheochromocytoma (PC12) cells ceased to proliferate and extend neurites. 3. The arrest of CLN-treated PC12 cells in the G1 phase of the cell cycle was due to an increase in the phosphorylation of p53 at serine(15) (p53ser15) and expression of p21WAF1. PC12 cells treated with inhibitory oligonucleotides to p53 lacked p53ser15 and p21WAF1 expression, and did not show morphological changes after CLN exposure. Transfection with inhibitory oligonucleotides to p21WAF1 had no effect on p53 activation; however, cells failed to arrest or extend neurites. An oligonucleotide inhibiting luciferase expression had no effect on CLN-mediated p53 activation, p21WAF1 expression, growth arrest, or neurite outgrowth. 4. We conclude that CLN induces delicate cassettes of signaling pathways common to cell proliferation and differentiation, and mediates activities that are similar to those of hormones and neurotrophins, leading to neurite outgrowth.
Collapse
Affiliation(s)
- Attila Bacsi
- Department of Microbiology and Immunology, University of Texas Medical Branch at Galveston, Galveston, TX 77555, USA
| | | | | | | | | |
Collapse
|
31
|
Hur J, Lee P, Kim J, Kim AJ, Kim H, Kim SY. Induction of nerve growth factor by butanol fraction of Liriope platyphylla in C6 and primary astrocyte cells. Biol Pharm Bull 2005; 27:1257-60. [PMID: 15305032 DOI: 10.1248/bpb.27.1257] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Liriope platyphylla (LP) has been used as a tonic, antitussive, and expectorant in Korea for many years. In this study, we found that the buthanol fraction of Liriope platyphylla (BLP)-conditioned media of C6 and primary astrocyte induced the neurite outgrowth of PC12 cells, and that the effect was reversed by addition of nerve growth factor (NGF)-antibody and GF109203X, an inhibitor of protein kinase (PKC). Furthermore, we demonstrated that BLP increased the expression and secretion of NGF. GF109203X also decreased NGF expression in C6 cells. Taken together, our results suggest that astroglial NGF enhanced by BLP in a PKC-dependent pathway contributed to the induction of neurite outgrowth of PC12 cells.
Collapse
Affiliation(s)
- Jinyoung Hur
- Graduate School of East-West Medical Science, Kyung Hee University, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
32
|
Schimmelpfeng J, Weibezahn KF, Dertinger H. Quantification of NGF-dependent neuronal differentiation of PC-12 cells by means of neurofilament-L mRNA expression and neuronal outgrowth. J Neurosci Methods 2004; 139:299-306. [PMID: 15488244 DOI: 10.1016/j.jneumeth.2004.05.010] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/06/2003] [Revised: 04/29/2004] [Accepted: 05/17/2004] [Indexed: 11/27/2022]
Abstract
We demonstrate that the degree of neuronal development of PC-12 cell differentiation can be quantified by the expression of neurofilament-L (NF-L) mRNA, when an optimal concentration of NGF (50 ng/ml) is used. During the first 7 days of NGF treatment, the relative amount of NF-L mRNA was found to increase continuously and to correlate with the outgrowth of neurites in a statistically significant way. Thus, mRNA expression is, under these conditions, a suitable means for reliably monitoring the differentiation of PC-12 cells as early as after 3 days of NGF treatment. The results obtained with 5 ng/ml NGF differ from those with 50 ng/ml: during the first 3 days of NGF treatment, neuronal outgrowth was less than with 50 ng/ml, although the NF-L mRNA levels did not depend significantly on NGF concentration. Beyond day 3, NF-L mRNA levels did not increase further at 5 ng/ml as opposed to 50 ng/ml NGF. These differences point to different signal transduction processes involved in neuronal differentiation at high and low NGF concentration. Expression of NF-L protein in response to NGF treatment was also demonstrated. In summary, our results stress that stable and sustained differentiation of PC-12 cells can only be achieved with 50 ng/ml NGF.
Collapse
Affiliation(s)
- J Schimmelpfeng
- Forschungszentrum Karlsruhe, Institute for Medical Engineering and Biophysics, P.O. Box 3640, D-76021 Karlsruhe, Germany.
| | | | | |
Collapse
|
33
|
Beaujean D, Rosenbaum C, Müller HW, Willemsen JJ, Lenders J, Bornstein SR. Combinatorial code of growth factors and neuropeptides define neuroendocrine differentiation in PC12 cells. Exp Neurol 2004; 184:348-58. [PMID: 14637105 DOI: 10.1016/j.expneurol.2003.07.007] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Adrenal chromaffin cells constitute one of the first cell types to have been defined as a neuroendocrine cell type. Since they produce dopamine, these cells have been proposed for the treatment of neuronal deficits in human Parkinson's disease. However, the factors involved in the development of chromaffin cells are still poorly understood. Based on recent insights from stem cell research, we decided to study the role of extracellular matrices, growth factors and neuropeptides on the neuroendocrine differentiation in a serum-free medium of PC12 cells. Employing immunohistochemistry, quantitative PCR and HPLC analysis, neuroendocrine differentiation was determined by evaluating neurite outgrowth, catecholamine biosynthesis and release as well as neuropeptide and vesicular protein mRNA expression. The combination of bFGF, NGF and PACAP could prevent the inhibition of neurite process development induced by dexamethasone in PC12 cells cultured on ECM. Whereas glucocorticoids were essential in the regulation of enzymes of catecholamine biosynthesis and metabolism, growth factors and PACAP were more efficient in inducing neuropeptide and chromogranin B expression as well as release of dopamine and 3-methoxytyramine. Therefore, in addition to glucocorticoids, chromaffin cells need a gradient of matrix, growth factors, and neuropeptides to develop the full functional phenotype of a neuroendocrine cell.
Collapse
Affiliation(s)
- Delphine Beaujean
- Department of Endocrinology, University of Düsseldorf, 40225, Düsseldorf, Germany.
| | | | | | | | | | | |
Collapse
|
34
|
Pappas TC, Decorti F, Macdonald NJ, Neet KE, Taglialatela G. Tumour necrosis factor-alpha- vs. growth factor deprivation-promoted cell death: different receptor requirements for mediating nerve growth factor-promoted rescue. Aging Cell 2003; 2:83-92. [PMID: 12882321 DOI: 10.1046/j.1474-9728.2003.00039.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
Physiological and pathological aging of the central nervous system (CNS) is characterized by functional neuronal impairments which may lead to perturbed cell homeostasis and eventually to neuronal death. Many toxic events may underlie age-related neurodegeneration. These include the effects of beta amyloid, Tau and mutated presenilin proteins, free radicals and oxidative stress, pro-inflammatory cytokines and lack of growth factor support, which can be individually or collectively involved. Taken individually, these toxicants can induce very diverse cell responses, thus requiring individually targeted corrective interventions upstream of common cell death (apoptotic) pathways. Recent preliminary evidence suggests that the pro-inflammatory cytokine tumour necrosis factor alpha (TNFalpha) and growth factor withdrawal can both activate a common apoptotic pathway in nerve growth factor (NGF)-responsive PC12 cells involving caspase 3, albeit through very distinct upstream pathways: the former through active signalling and the latter through passive or lack of survival signalling. Here, we show that NGF can rescue PC12 cells from both growth factor withdrawal- and TNFalpha-promoted cell death. However, NGF rescue from growth factor withdrawal requires NGF signalling through the high-affinity tyrosine kinase receptor (TrkA), while NGF rescue from TNFalpha-promoted cell death requires NGF signalling through the low-affinity p75NTR receptor. These results strengthen the idea that prevention of age- or pathology-associated neurodegeneration may require varied molecular approaches reflecting the diversity of the toxicants involved, possibly acting simultaneously.
Collapse
Affiliation(s)
- Todd C Pappas
- Center for Biomedical Engineering, The University of Texas Medical Branch at Galveston, Galveston, TX 77555 -1043, USA
| | | | | | | | | |
Collapse
|
35
|
Mariggiò MA, Guarnieri S, Mariggiò S, Morabito C, Gianfranceschi GL, Fanò G. N-CAM expression and localization in PC12 cells modulated by extracellular peptides. Peptides 2002; 23:2151-61. [PMID: 12535693 DOI: 10.1016/s0196-9781(02)00258-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/09/2023]
Abstract
The neural cell adhesion molecules (N-CAMs) play an important role in mediating cell-cell interactions in the nervous system. Different isoforms of these membrane proteins are involved in the formation of the neuronal network and in the dynamic phases of neuronal plasticity. We studied the early stages of the pseudo neuronal differentiation of PC12 cells induced by a class of small acidic peptides capable of modulating gene expression in these cells. The data presented here indicate that peptides with specific sequences induce an increase in N-CAM mRNA expression and protein translocation to the plasma membrane to a comparable degree as NGF.
Collapse
Affiliation(s)
- Maria A Mariggiò
- Dipartimento di Scienze del Farmaco, Laboratorio di Fisiologia Cellulare, Università G. d'Annunzio, 66013 Chieti, Italy.
| | | | | | | | | | | |
Collapse
|
36
|
Luo HR, Saiardi A, Nagata E, Ye K, Yu H, Jung TS, Luo X, Jain S, Sawa A, Snyder SH. GRAB: a physiologic guanine nucleotide exchange factor for Rab3A, which interacts with inositol hexakisphosphate kinase. Neuron 2001; 31:439-51. [PMID: 11516400 DOI: 10.1016/s0896-6273(01)00384-1] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Diphosphoinositol-pentakisphosphate (InsP7) and bis-diphosphoinositol tetrakisphosphate (InsP8) possess pyrophosphate bonds. InsP7 is formed from inositol hexakisphosphate (InsP6) by recently identified InsP6 kinases designated InsP6K1 and InsP6K2. We now report the identification, cloning, and characterization of a novel protein, GRAB (guanine nucleotide exchange factor for Rab3A), which interacts with both InsP6K1 and Rab3A, a Ras-like GTPase that regulates synaptic vesicle exocytosis. GRAB is a physiologic GEF (guanine nucleotide exchange factor) for Rab3A. Consistent with a role of Rab3A in synaptic vesicle exocytosis, GRAB regulates depolarization-induced release of dopamine from PC12 cells and nicotinic agonist-induced hGH release from bovine adrenal chromaffin cells. The association of InsP6K1 with GRAB fits with a role for InsP7 in vesicle exocytosis.
Collapse
Affiliation(s)
- H R Luo
- Department of Neuroscience, School of Medicine, Johns Hopkins University, 725 N. Wolfe Street, Baltimore, MD 21205, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
37
|
Macdonald NJ, Decorti F, Pappas TC, Taglialatela G. Cytokine/neurotrophin interaction in the aged central nervous system. J Anat 2000; 197 Pt 4:543-51. [PMID: 11197527 PMCID: PMC1468169 DOI: 10.1046/j.1469-7580.2000.19740543.x] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Age-associated neurodegenerative diseases such as Alzheimer's disease are characterised by neuronal impairment that leads to cognitive deficits. As certain affected neurons depend on trophic factors such as neurotrophins (NTs), impairment in NT function has been suggested to be a component of neuronal damage associated with such disorders. Age-related neurodegenerative diseases are also characterised by high levels of proinflammatory cytokines such as tumour necrosis factor alpha (TNFalpha) in the CNS. Because TNFalpha receptors and certain NT receptors share a high degree of homology and are capable of activating similar signalling pathways, one possibility is that altered cytokine levels may affect NT function in the aged or diseased CNS. Here we wish briefly to review the evidence suggesting a role for cytokine and NT in the onset of age-associated neurodegenerative diseases. We propose that cytokine/NT interactions may alter neuronal homeostasis, thus possibly contributing to some of the neuronal degeneration occurring during such age-associated CNS diseases.
Collapse
Affiliation(s)
- N J Macdonald
- Department of Anatomy and Neuroscience and Marine Biomedical Institute University of Texas Medical Branch at Galveston, 77555-0652, USA
| | | | | | | |
Collapse
|
38
|
Straub M, Lodemann P, Holroyd P, Jahn R, Hell SW. Live cell imaging by multifocal multiphoton microscopy. Eur J Cell Biol 2000; 79:726-34. [PMID: 11089921 DOI: 10.1078/0171-9335-00105] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Multifocal multiphoton microscopy (MMM) permits parallel multiphoton excitation by scanning an array of high numerical aperture foci across a plane in the sample. MMM is particularly suitable for live cell investigations since it combines advantages of standard multiphoton microscopy such as optical sectioning and suppression of out-of-focus phototoxicity with high recording speeds. Here we describe several applications of MMM to live cell imaging using the neuroendocrine cell line PC12 and bovine chromaffin cells. Stainings were performed with the acidophilic dye acridine orange and the lipophilic dyes FM1-43 and Fast DiA as well as by transfection of the cells with GFP. In both bovine chromaffin and PC12 cells structural elements of nuclear chromatin and the 3-D distribution of acidic organelles inside the cells were visualized. In PC12 cells differentiated by nerve growth factor examples of neurites were monitored. Stainings of membranes were used to reconstruct the morphology of cells and neurites in three dimensions by volume-rendering and by isosurface plots. 3-D reconstructions were composed from stacks of about 50 images each with a diameter of 30-100 microm that were acquired within a few seconds. We conclude that MMM proves to be a technically simple and very effective method for fast 3-D live cell imaging at high resolution.
Collapse
Affiliation(s)
- M Straub
- High Resolution Optical Microscopy Group, Max-Planck-Institute for Biophysical Chemistry, Göttingen, Germany.
| | | | | | | | | |
Collapse
|
39
|
Farooqui AA, Litsky ML, Farooqui T, Horrocks LA. Inhibitors of intracellular phospholipase A2 activity: their neurochemical effects and therapeutical importance for neurological disorders. Brain Res Bull 1999; 49:139-53. [PMID: 10435777 DOI: 10.1016/s0361-9230(99)00027-1] [Citation(s) in RCA: 97] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Intracellular phospholipases A2 (PLA2) are a diverse group of enzymes with a growing number of members. These enzymes hydrolyze membrane phospholipids into fatty acid and lysophospholipids. These lipid products may serve as intracellular second messengers or can be further metabolized to potent inflammatory mediators, such as eicosanoids and platelet-activating factors. Several inhibitors of nonneural intracellular PLA2 have been recently discovered. However, nothing is known about their neurochemical effects, mechanism of action or toxicity in human or animal models of neurological disorders. Elevated intracellular PLA2 activities, found in neurological disorders strongly associated with inflammation and oxidative stress (ischemia, spinal cord injury, and Alzheimer's disease), can be treated with specific, potent and nontoxic inhibitors of PLA2 that can cross blood-brain barrier without harm. Currently, potent intracellular PLA2 inhibitors are not available for clinical use in human or animal models of neurological disorders, but studies on this interesting topic are beginning to emerge. The use of nonspecific intracellular PLA2 inhibitors (quinacrine, heparin, gangliosides, vitamin E) in animal model studies of neurological disorders in vivo has provided some useful information on tolerance, toxicity, and effectiveness of these compounds.
Collapse
Affiliation(s)
- A A Farooqui
- Department of Medical Biochemistry, The Ohio State University, Columbus 43210, USA.
| | | | | | | |
Collapse
|
40
|
Pan Z, Sampath D, Jackson G, Werrbach-Perez K, Perez-Polo R. Nerve growth factor and oxidative stress in the nervous system. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 1997; 429:173-93. [PMID: 9413574 DOI: 10.1007/978-1-4757-9551-6_13] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Affiliation(s)
- Z Pan
- Department of Human Biological Chemistry and Genetics, University of Texas Medical Branch at Galveston 77555-0652, USA
| | | | | | | | | |
Collapse
|
41
|
Grimes ML, Beattie E, Mobley WC. A signaling organelle containing the nerve growth factor-activated receptor tyrosine kinase, TrkA. Proc Natl Acad Sci U S A 1997; 94:9909-14. [PMID: 9275225 PMCID: PMC23291 DOI: 10.1073/pnas.94.18.9909] [Citation(s) in RCA: 145] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/1997] [Accepted: 07/03/1997] [Indexed: 02/05/2023] Open
Abstract
The topology of signal transduction is particularly important for neurons. Neurotrophic factors such as nerve growth factor (NGF) interact with receptors at distal axons and a signal is transduced by retrograde transport to the cell body to ensure survival of the neuron. We have discovered an organelle that may account for the retrograde transport of the neurotrophin signal. This organelle is derived from endocytosis of the receptor tyrosine kinase for NGF, TrkA. In vitro reactions containing semi-intact PC12 cells and ATP were used to enhance recovery of a novel organelle: small vesicles containing internalized NGF bound to activated TrkA. These vesicles were distinct from clathrin coated vesicles, uncoated primary endocytic vesicles, and synaptic vesicles, and resembled transport vesicles in their sedimentation velocity. They contained 10% of the total bound NGF and almost one-third of the total tyrosine phosphorylated TrkA. These small vesicles are compelling candidates for the organelles through which the neurotrophin signal is conveyed down the axon.
Collapse
Affiliation(s)
- M L Grimes
- Department of Biochemistry, Massey University, Palmerston North, New Zealand.
| | | | | |
Collapse
|
42
|
Taglialatela G, Robinson R, Perez-Polo JR. Inhibition of nuclear factor kappa B (NF?B) activity induces nerve growth factor-resistant apoptosis in PC12 cells. J Neurosci Res 1997. [DOI: 10.1002/(sici)1097-4547(19970115)47:2<155::aid-jnr4>3.0.co;2-e] [Citation(s) in RCA: 137] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
|
43
|
Lyons WE, George EB, Dawson TM, Steiner JP, Snyder SH. Immunosuppressant FK506 promotes neurite outgrowth in cultures of PC12 cells and sensory ganglia. Proc Natl Acad Sci U S A 1994; 91:3191-5. [PMID: 7512727 PMCID: PMC43541 DOI: 10.1073/pnas.91.8.3191] [Citation(s) in RCA: 212] [Impact Index Per Article: 7.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
The immunosuppressant drug FK506 acts by binding to receptor proteins, FK506-binding proteins (FKBPs), which in turn can bind to and regulate a Ca(2+)-dependent phosphatase, calcineurin, and a Ca2+ release channel, the ryanodine receptor. Based on our findings in regeneration models that levels of FKBPs during neural regeneration parallel those of growth-associated protein GAP43, a calcineurin substrate that regulates neurite extension, we examined effects of FK506 in PC12 rat pheochromocytoma cells and in rat sensory ganglia. FK506 enhances neurite outgrowth in both systems by increasing sensitivity to nerve growth factor. Blockade of FK506 actions in sensory ganglia by rapamycin, an FK506 antagonist, establishes that these effects involve FKBPs. Rapamycin itself stimulates neurite outgrowth in PC12 cells. These drug effects are detected at subnanomolar concentrations, suggesting therapeutic application in diseases involving neural degeneration.
Collapse
Affiliation(s)
- W E Lyons
- Division of Toxicological Sciences, Johns Hopkins University School of Medicine, Baltimore, MD 21205
| | | | | | | | | |
Collapse
|
44
|
Blackman CF, Blanchard JP, Benane SG, House DE. Empirical test of an ion parametric resonance model for magnetic field interactions with PC-12 cells. Bioelectromagnetics 1994; 15:239-60. [PMID: 8074739 DOI: 10.1002/bem.2250150307] [Citation(s) in RCA: 80] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023]
Abstract
A companion paper describes a predictive ion parametric resonance (IPR) model of magnetic field interactions with biological systems based on a selective relation between the ratio of the flux density of the static magnetic field to the AC magnetic field and the charge-to-mass ratio of ions of biological relevance. Previous studies demonstrated that nerve growth factor (NGF)-stimulated neurite outgrowth (NO) in PC-12 cells can be inhibited by exposure to magnetic fields as a function of either magnetic field flux density or AC magnetic field frequency. The present work examines whether the PC-12 cell response to magnetic fields is consistent with the quasi-periodic, resonance-based predictions of the IPR model. We tested changes in each of the experimentally controllable variables [flux densities of the parallel components of the AC magnetic field (Bac) and the static magnetic field (Bdc) and the frequency of the AC magnetic field] over a range of exposure conditions sufficient to determine whether the IPR model is applicable. A multiple-coil exposure system independently controlled each of these critical quantities. The perpendicular static magnetic field was controlled to less than 2 mG for all tests. The first set of tests examined the NO response in cells exposed to 45 Hz Bac from 77 to 468 mG(rms) at a Bdc of 366 mG. Next, we examined an off-resonance condition using 20 mG Bdc with a 45 Hz AC field across a range of Bac between 7.9 and 21 mG(rms). Finally, we changed the AC frequency to 25 Hz, with a corresponding change in Bdc to 203 mG (to tune for the same set of ions as in the first test) and a Bac range from 78 to 181 mG(rms). In all cases the observed responses were consistent with predictions of the IPR model. These experimental results are the first to support in detail the validity of the fundamental relationships embodied in the IPR model.
Collapse
Affiliation(s)
- C F Blackman
- Health Effects Research Laboratory, U.S. Environmental Protection Agency, Research Triangle Park, North Carolina 27711
| | | | | | | |
Collapse
|
45
|
de la Torre JC, Rall G, Oldstone C, Sanna PP, Borrow P, Oldstone MB. Replication of lymphocytic choriomeningitis virus is restricted in terminally differentiated neurons. J Virol 1993; 67:7350-9. [PMID: 8230458 PMCID: PMC238199 DOI: 10.1128/jvi.67.12.7350-7359.1993] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
We have investigated the replication of lymphocytic choriomeningitis virus (LCMV) before and after the nerve growth factor (NGF)-induced transdifferentiation of PC12 cells from the chromaffin to the neuron-like phenotype. Untreated and NGF-treated cells were equally susceptible to LCMV infection; however, the viral yield was found to be 1,000-fold lower in NGF-differentiated PC12 cells. The reduced viral yield correlated with restricted LCMV replication and transcription within the infected cell, which was not caused by the lack of cell proliferation in the NGF-treated cells but rather was related to the induction or changes in expression levels of specific gene product(s) associated with the cell commitment to a neuronal phenotype. The return to the chromaffin phenotype after withdrawal of NGF restored normal LCMV yields as well as levels of viral replication and transcription. The finding of reduced viral replication in terminally differentiated neuronal cells has important implications for understanding the mechanism by which neurotropic viruses, such as LCMV, are able to establish a long-term persistent infection in the central nervous system in the absence of severe pathological changes.
Collapse
Affiliation(s)
- J C de la Torre
- Department of Neuropharmacology, Scripps Research Institute, La Jolla, California 92037
| | | | | | | | | | | |
Collapse
|
46
|
Hirsch DB, Steiner JP, Dawson TM, Mammen A, Hayek E, Snyder SH. Neurotransmitter release regulated by nitric oxide in PC-12 cells and brain synaptosomes. Curr Biol 1993; 3:749-54. [PMID: 15335838 DOI: 10.1016/0960-9822(93)90022-g] [Citation(s) in RCA: 75] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/1993] [Revised: 09/13/1993] [Accepted: 10/14/1993] [Indexed: 10/26/2022]
Abstract
BACKGROUND Nitric oxide is a messenger molecule of the nervous system, which is produced by the enzyme nitric oxide synthase, which may regulate cyclic guanosine monophosphate levels and which has been implicated in the control of neurotransmitter release. PC-12 pheochromocytoma cells differentiate to form neuronal cells in culture when they are exposed to nerve growth factor. The levels of cyclic guanosine monophosphate in the cells and their ability to release acetylcholine in response to K(+)-depolarization are both maximal after eight days of treatment with nerve growth factor. We set out to assess a possible role for nitric oxide in the processes that occur in differentiating PC-12 cells. RESULTS Nitric oxide synthase is first evident in differentiating PC-12 cells eight days after beginning treatment with nerve growth factor, coinciding with the marked increase in K(+)-depolarization-induced release of acetylcholine. The release of both acetylcholine and dopamine in response to K(+)-depolarization is blocked by inhibitors of nitric oxide synthase and by hemoglobin, which binds nitric oxide. Providing l-arginine, a precursor required for nitric oxide synthesis, reverses the effects of the inhibitors. In synaptosomal preparations from the corpus striatum, inhibitors of nitric oxide synthase prevent the release of glutamate in response to the glutamate derivative N-methyl-d-aspartate but not in response to K(+)-depolarization. CONCLUSION Nitric oxide may mediate the release of acetylcholine and dopamine in response to K(+)-depolarization in PC-12 cells and the release of glutamate in response to N-methyl-d-aspartate in striatal synaptosomes. Nitric oxide synthase expression is induced after eight days of treating PC-12 cells with nerve growth factor, coinciding with a marked enhancement of the release of neurotransmitters in response to K(+)-depolarization.
Collapse
Affiliation(s)
- D B Hirsch
- Departments of Neuroscience, Pharmacology and Molecular Sciences, Johns Hopkins University School of Medicine, Baltimore, Maryland 21205, USA
| | | | | | | | | | | |
Collapse
|
47
|
Volonté C. Dexamethasone abolishes the activation by nerve growth factor of protein kinase N: effects of nerve growth factor and dexamethasone on protein kinase N. Neurosci Lett 1993; 159:119-22. [PMID: 8264951 DOI: 10.1016/0304-3940(93)90813-z] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Protein kinase N (PKN) is a basic 45-47 kDa serine/threonine protein kinase activated by NGF and several other factors in PC12 cells and other cell types. It is inhibited in vitro by purine analogs. In this work, we further characterize the modulation of PKN activity following exposure of PC12 cells to dexamethasone or NGF, which respectively direct these cells towards the chromaffin- or neuron-like phenotype. We show here that the two factors elicit opposite effects on the activation of the kinase: dexamethasone inhibits while NGF stimulates the basal level of PKN activity. Simultaneous addition to the cells of the factors causes no variation from basal kinase activity. Addition of dexamethasone not to the cells, but to partially purified PKN during the kinase assay, causes no modulation of the enzyme.
Collapse
Affiliation(s)
- C Volonté
- Department of Pathology, College of Physicians and Surgeons, Columbia University, New York, NY 10032
| |
Collapse
|
48
|
Akaike N, Furukawa K, Kogure K. Rolipram enhances the development of voltage-dependent Ca2+ current and serotonin-induced current in rat pheochromocytoma cells. Brain Res 1993; 620:58-63. [PMID: 8402199 DOI: 10.1016/0006-8993(93)90270-w] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
The effects of chronic treatment (6-8 days) with a phosphodiesterase inhibitor, rolipram, on the expression of voltage-dependent Ca2+ channels, nicotinic acetylcholine (ACh) receptors and 5-hydroxytryptamine (5-HT) receptors were investigated in PC12 cells. The results were compared with the effects of nerve growth factor (NGF), 8-bromo-cyclic AMP (8-Br-cAMP) and phorbol 12-myristate 13-acetate (PMA). In the morphological study rolipram, at a high concentration (100 microM) induced the extension of neurites. A similar result was obtained in 8-Br-cAMP (1 mM)-treated cells. Rolipram, at a low concentration (10 microM) or PMA (10(-7) M) did not induce obvious morphological change. NGF (100 ng/ml) induced the extension of long neurites and the formation of neural networks. Rolipram (100 microM) increased the current density (pA/pF) of voltage dependent Ca2+ current (ICa). Both NGF and 8-Br-cAMP also increased the current density of ICa, whereas PMA did not. NGF increased the current density of the nicotinic ACh response whereas rolipram, 8-Br-cAMP and PMA decreased. Rolipram (100 microM), NGF (100 ng/ml), and 8-Br-cAMP (1 mM) increased the current density of the 5-HT response whereas the effect of PMA (100 nM) was slight. The results suggest that rolipram is able to contribute to the neuronal development by increasing intracellular cAMP as well as 8-Br-cAMP. Consequently, rolipram behaves like a neurotrophic factor in cultured PC12 cells.
Collapse
Affiliation(s)
- N Akaike
- Department of Neurophysiology Tohoku University School of Medicine, Sendai, Japan
| | | | | |
Collapse
|
49
|
Volonté C, Loeb DM, Greene LA. A purine analog-sensitive protein kinase activity associates with Trk nerve growth factor receptors. J Neurochem 1993; 61:664-72. [PMID: 7687657 DOI: 10.1111/j.1471-4159.1993.tb02171.x] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Previous studies showed that purine analogs block with varying efficiency and specificity certain effects of nerve growth factor (NGF) on PC12 cells. These compounds also inhibit protein kinase activities. The analog 6-thioguanine has thus far been shown to inhibit only protein kinase N, an NGF-activated protein kinase, whereas 2-aminopurine also blocks other kinases. In the present study, immunoprecipitates of Trk NGF receptors from PC12 cells (+/- NGF treatment) were assayed for protein kinase activity by using the substrates myelin basic protein and histone HF1 under phosphorylating conditions optimal for protein kinase N and in the presence or absence of purine analogs. Activity was detected and approximately 50-80% was inhibited by these compounds. The purine analog-sensitive activity was maximally stimulated by NGF within 5 min, was partially decreased by 10 min, and still remained over basal levels after 15 h of NGF treatment. Analysis of myelin basic protein phosphorylated by anti-Trk immunoprecipitates revealed an NGF-stimulated increase in phosphothreonine and phosphotyrosine. Phosphorylation of threonine, but not of tyrosine residues, was inhibited by 6-thioguanine, which therefore inhibits a serine/threonine kinase associated with NGF receptor rather than the receptor kinase itself. Neither 2-aminopurine nor 6-thioguanine inhibited the NGF-dependent induction of Trk-associated kinase activity. Our findings thus indicate association of a purine analog-sensitive serine/threonine protein kinase activity with Trk NGF receptors.
Collapse
Affiliation(s)
- C Volonté
- Department of Pathology, College of Physicians and Surgeons, Columbia University, New York, New York
| | | | | |
Collapse
|
50
|
Ford TA, Mueller GP. Induction of peptidylglycine alpha-hydroxylating monooxygenase activity by nerve growth factor in PC12 cells. J Mol Neurosci 1993; 4:97-105. [PMID: 8217523 DOI: 10.1007/bf02782122] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Pheochromocytoma PC12 cells grown in the presence of nerve growth factor (NGF) undergo marked neuronal differentiation. During this process gene expression is altered, resulting in the activation of genes specific for neuronal properties, including the gene encoding neuropeptide Y (NPY). Here we sought to determine whether NGF also induces the activity of peptidylglycine alpha-hydroxylating monooxygenase (PHM) (EC1.4.17.3). PHM catalyzes the rate limiting step in the formation of alpha-amidated NPY from its glycine extended precursor, a posttranslational modification essential for biologic activity. PC12 cells were grown with or without NGF and assayed for PHM activity under optimal conditions. Whole cell extracts, medium and soluble and membrane bound fractions were assayed; total cellular PHM activity was found to be primarily membrane bound (fivefold greater than in soluble) and very little activity was released into the medium. Compared to control cells, PHM activity was increased significantly by NGF by 24 h but not before 4 h exposure. Through kinetic analysis, it was determined that the NGF-induction of PHM was a result of an increase in Vmax with no change in Km. It was found that the glucocorticoid, dexamethasone (DEX), decreased basal PHM activity and prevented its induction by NGF. Cotreatment with DEX for up to 7 d, however, did not dramatically alter the pronounced changes in cell morphology that occurred in response to NGF. These findings indicate that NGF and glucocorticoids exert reciprocal control over the activity of PHM in PC12 cells. As such, the process of differentiation in PC12 cells is a model for studying the mechanisms that coordinate the expression and activity of peptide processing enzymes with the regulation of their substrates and products.
Collapse
Affiliation(s)
- T A Ford
- Department of Physiology, Uniformed Services University of the Health Sciences, Bethesda, MD 20814-4799
| | | |
Collapse
|