1
|
Duarte-Silva AT, Ximenes LGR, Guimarães-Souza M, Domith I, Paes-de-Carvalho R. Chemical signaling in the developing avian retina: Focus on cyclic AMP and AKT-dependent pathways. Front Cell Dev Biol 2022; 10:1058925. [PMID: 36568967 PMCID: PMC9780464 DOI: 10.3389/fcell.2022.1058925] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2022] [Accepted: 11/28/2022] [Indexed: 12/13/2022] Open
Abstract
Communication between developing progenitor cells as well as differentiated neurons and glial cells in the nervous system is made through direct cell contacts and chemical signaling mediated by different molecules. Several of these substances are synthesized and released by developing cells and play roles since early stages of Central Nervous System development. The chicken retina is a very suitable model for neurochemical studies, including the study of regulation of signaling pathways during development. Among advantages of the model are its very well-known histogenesis, the presence of most neurotransmitter systems found in the brain and the possibility to make cultures of neurons and/or glial cells where many neurochemical functions develop in a similar way than in the intact embryonic tissue. In the chicken retina, some neurotransmitters or neuromodulators as dopamine, adenosine, and others are coupled to cyclic AMP production or adenylyl cyclase inhibition since early stages of development. Other substances as vitamin C and nitric oxide are linked to the major neurotransmitter glutamate and AKT metabolism. All these different systems regulate signaling pathways, including PKA, PKG, SRC, AKT and ERK, and the activation of the transcription factor CREB. Dopamine and adenosine stimulate cAMP accumulation in the chick embryo retina through activation of D1 and A2a receptors, respectively, but the onset of dopamine stimulation is much earlier than that of adenosine. However, adenosine can inhibit adenylyl cyclase and modulate dopamine-dependent cAMP increase since early developmental stages through A1 receptors. Dopamine stimulates different PKA as well as EPAC downstream pathways both in intact tissue and in culture as the CSK-SRC pathway modulating glutamate NMDA receptors as well as vitamin C release and CREB phosphorylation. By the other hand, glutamate modulates nitric oxide production and AKT activation in cultured retinal cells and this pathway controls neuronal survival in retina. Glutamate and adenosine stimulate the release of vitamin C and this vitamin regulates the transport of glutamate, activation of NMDA receptors and AKT phosphorylation in cultured retinal cells. In the present review we will focus on these reciprocal interactions between neurotransmitters or neuromodulators and different signaling pathways during retinal development.
Collapse
Affiliation(s)
- A. T. Duarte-Silva
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - L. G. R. Ximenes
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - M. Guimarães-Souza
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - I. Domith
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil
| | - R. Paes-de-Carvalho
- Program of Neurosciences, Institute of Biology, Fluminense Federal University, Niterói, Brazil,Department of Neurobiology, Institute of Biology, Fluminense Federal University, Niterói, Brazil,*Correspondence: R. Paes-de-Carvalho,
| |
Collapse
|
2
|
The Role of PKC and HIF-1 and the Effect of Traditional Chinese Medicinal Compounds on Cerebral Ischemia-Reperfusion Injury. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2022; 2022:1835898. [PMID: 35265143 PMCID: PMC8898791 DOI: 10.1155/2022/1835898] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/03/2021] [Revised: 01/26/2022] [Accepted: 02/07/2022] [Indexed: 11/25/2022]
Abstract
Neuronal death occurs during cerebral ischemia. However, when hemoperfusion and oxygen supply are resumed to the ischemic focus of the brain tissue, the brain tissue damage is further aggravated, resulting in cerebral ischemia-reperfusion injury (CIRI) to the patients. Protein kinase C (PKC) plays an important role in CIRI. Through the IP3/DAG/Ca2+ signaling pathway, it promotes the influx of calcium ions in neurons and causes calcium overload, which aggravates the damage. At the same time, when brain cells are hypoxic, hypoxia-inducible factor-1 (HIF-1) is expressed, which regulates the expression of Bcl-2 and Bax through the PI3K/Akt signaling pathway and reduces nerve cell injury. It also fights hypoxic-ischemic injury by increasing the production of vascular endothelial growth factor (VEGF) to promote blood vessel formation. The PKC and HIF-1 signaling pathways are also linked to CIRI. HIF-1 activates the PKC and ERK pathways via the upregulation of VEGF, leading to increased Cx43 phosphorylation and dysfunction and aggravating CIRI. Existing studies have shown that certain traditional Chinese medicine (TCM) compounds regulate the PKC and HIF-1 signaling pathways and alleviate CIRI. These compounds downregulate the PKC and the activity of the PKC-related signaling pathways to alleviate CIRI. They can also promote the expression of HIF-1, increase the content of VEGF in ischemic tissues to promote the generation of blood vessels, and improve microcirculation. TCM compounds can inhibit the cascade of reactions underlying disease occurrence and development by targeting multiple components using different herbal formulations to improve the structural and material changes in the brain cells, which alleviate CIRI and protect the brain tissue. This study briefly describes the role of PKC and HIF-1, their relationship in CIRI, and the effect of TCM on them.
Collapse
|
3
|
Sheng R, Chen JL, Qin ZH. Cerebral conditioning: Mechanisms and potential clinical implications. BRAIN HEMORRHAGES 2021. [DOI: 10.1016/j.hest.2021.08.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
|
4
|
Cilleros-Mañé V, Just-Borràs L, Polishchuk A, Durán M, Tomàs M, Garcia N, Tomàs JM, Lanuza MA. M 1 and M 2 mAChRs activate PDK1 and regulate PKC βI and ε and the exocytotic apparatus at the NMJ. FASEB J 2021; 35:e21724. [PMID: 34133802 DOI: 10.1096/fj.202002213r] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2020] [Revised: 05/07/2021] [Accepted: 05/24/2021] [Indexed: 01/14/2023]
Abstract
Neuromuscular junctions (NMJ) regulate cholinergic exocytosis through the M1 and M2 muscarinic acetylcholine autoreceptors (mAChR), involving the crosstalk between receptors and downstream pathways. Protein kinase C (PKC) regulates neurotransmission but how it associates with the mAChRs remains unknown. Here, we investigate whether mAChRs recruit the classical PKCβI and the novel PKCε isoforms and modulate their priming by PDK1, translocation and activity on neurosecretion targets. We show that each M1 and M2 mAChR activates the master kinase PDK1 and promotes a particular priming of the presynaptic PKCβI and ε isoforms. M1 recruits both primed-PKCs to the membrane and promotes Munc18-1, SNAP-25, and MARCKS phosphorylation. In contrast, M2 downregulates PKCε through a PKA-dependent pathway, which inhibits Munc18-1 synthesis and PKC phosphorylation. In summary, our results discover a co-dependent balance between muscarinic autoreceptors which orchestrates the presynaptic PKC and their action on ACh release SNARE-SM mechanism. Altogether, this molecular signaling explains previous functional studies at the NMJ and guide toward potential therapeutic targets.
Collapse
Affiliation(s)
- V Cilleros-Mañé
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - L Just-Borràs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - A Polishchuk
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - M Durán
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - M Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - N Garcia
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - J M Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| | - M A Lanuza
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Departament de Ciències Mèdiques Bàsiques, Universitat Rovira i Virgili, Reus, Spain
| |
Collapse
|
5
|
Zhang Y, Cao H, Qiu X, Xu D, Chen Y, Barnes GN, Tu Y, Gyabaah AT, Gharbal AHAA, Peng C, Cai J, Cai X. Neuroprotective Effects of Adenosine A1 Receptor Signaling on Cognitive Impairment Induced by Chronic Intermittent Hypoxia in Mice. Front Cell Neurosci 2020; 14:202. [PMID: 32733207 PMCID: PMC7363980 DOI: 10.3389/fncel.2020.00202] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2020] [Accepted: 06/09/2020] [Indexed: 12/18/2022] Open
Abstract
Obstructive sleep apnea-hypopnea syndrome (OSAHS) is a breathing disorder associated with cognitive impairment. However, the mechanisms leading to cognitive deficits in OSAHS remain uncertain. In this study, a mouse model of chronic intermittent hypoxia (CIH) exposures were applied for simulating the deoxygenation-reoxygenation events occurring in OSAHS. The conventional adenosine A1 receptor gene (A1R) knockout mice and the A1R agonist CCPA- or antagonist DPCPX-administrated mice were utilized to determine the precise function of A1R signaling in the process of OSAHS-relevant cognitive impairment. We demonstrated that CIH induced morphological changes and apoptosis in hippocampal neurons. Further, CIH blunted hippocampal long-term potentiation (LTP) and resulted in learning/memory impairment. Disruption of adenosine A1R exacerbated morphological, cellular, and functional damage induced by CIH. In contrast, activation of adenosine A1R signaling reduced morphological changes and apoptosis of hippocampal neurons, promoted LTP, and enhanced learning and memory. A1Rs may up-regulate protein kinase C (PKC) and its subtype PKC-ζ through the activation of Gα(i) improve spatial learning and memory disorder induced by CIH in mice. Taken together, A1R signaling plays a neuroprotective role in CIH-induced cognitive dysfunction and pathological changes in the hippocampus.
Collapse
Affiliation(s)
- Yichun Zhang
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Hongchao Cao
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China.,Department of Internal Medicine, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No. 2 Hospital), Ningbo, China
| | - Xuehao Qiu
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Danfen Xu
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Yifeng Chen
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Gregory N Barnes
- Department of Neurology, University of Louisville School of Medicine, Louisville, KY, United States.,Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, KY, United States
| | - Yunjia Tu
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | - Adwoa Takyiwaa Gyabaah
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| | | | - Chenlei Peng
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China.,Department of Internal Medicine, Hwa Mei Hospital, University of Chinese Academy of Sciences (Ningbo No. 2 Hospital), Ningbo, China
| | - Jun Cai
- Department of Pediatrics, Pediatric Research Institute, University of Louisville School of Medicine, Louisville, KY, United States
| | - Xiaohong Cai
- Department of Pediatrics, The Second Affiliated Hospital and Yuying Children's Hospital, Wenzhou Medical University, Wenzhou, China
| |
Collapse
|
6
|
PKCγ and PKCε are Differentially Activated and Modulate Neurotoxic Signaling Pathways During Oxygen Glucose Deprivation in Rat Cortical Slices. Neurochem Res 2019; 44:2577-2589. [PMID: 31541352 DOI: 10.1007/s11064-019-02876-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/18/2019] [Revised: 09/10/2019] [Accepted: 09/13/2019] [Indexed: 10/26/2022]
Abstract
Cerebral ischemia is known to trigger a series of intracellular events such as changes in metabolism, membrane function and intracellular transduction, which eventually leads to cell death. Many of these processes are mediated by intracellular signaling cascades that involve protein kinase activation. Among all the kinases activated, the serine/threonine kinase family, protein kinase C (PKC), particularly, has been implicated in mediating cellular response to cerebral ischemic and reperfusion injury. In this study, using oxygen-glucose deprivation (OGD) in acute cortical slices as an in vitro model of cerebral ischemia, I show that PKC family of isozymes, specifically PKCγ and PKCε are differentially activated during OGD. Detecting the expression and activation levels of these isozymes in response to different durations of OGD insult revealed an early activation of PKCε and delayed activation of PKCγ, signifying their roles in response to different durations and stages of ischemic stress. Specific inhibition of PKCγ and PKCε significantly attenuated OGD induced cytotoxicity, rise in intracellular calcium, membrane depolarization and reactive oxygen species formation, thereby enhancing neuronal viability. This study clearly suggests that PKC family of isozymes; specifically PKCγ and PKCε are involved in OGD induced intracellular responses which lead to neuronal death. Thus isozyme specific modulation of PKC activity may serve as a promising therapeutic route for the treatment of acute cerebral ischemic injury.
Collapse
|
7
|
Dang YF, Qiu TX, Song DW, Liu L. PMA-triggered PKCε activity enhances Nrf2-mediated antiviral response on fish rhabdovirus infection. FISH & SHELLFISH IMMUNOLOGY 2019; 87:871-878. [PMID: 30776542 DOI: 10.1016/j.fsi.2019.02.033] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/25/2018] [Revised: 02/12/2019] [Accepted: 02/15/2019] [Indexed: 06/09/2023]
Abstract
Viral infection is often accompanied with alteration of intracellular redox state, especially an imbalance between reactive oxygen species (ROS) production and antioxidant cellular defenses. The previous studies showed that an antioxidant cellular defense system, the transcription factor nuclear factor erythroid 2-related factor 2 (Nrf2), played an important role against spring viraemia of carp virus (SVCV) infection in fish. To further reveal the mediated mechanism that Nrf2 active state was affected by protein kinase C (PKC), here we evaluated SVCV replication in host cells by treated with a strong activator of PKC phorbol-12-myristate-13-acetate (PMA) and an inhibitor staurosporine. Our results showed that PMA significantly repressed SVCV replication and viral-induced apoptosis in Epithelioma papulosum cyprini (EPC) cell, suggesting that PKC may exhibit an anti-SVCV effect. Likewise, PMA resulted in a higher phosphorylation levels of PKCε rather than PKCα/β to participate in the activation of Nrf2, mainly involved in the activation of Nrf2 phosphorylation of Ser40 to favor Nrf2 translocation to nucleus. Furthermore, the data revealed that PMA up-regulated an antiviral response heme oxygenase-1 (HO1) gene expression that was confirmed as the key player against SVCV infection by HO1 specific siRNA. Overall, this study provided a new therapeutic target for the treatment of SVCV infection, and modulating PKC activity could be used for the prevention and treatment of SVCV.
Collapse
Affiliation(s)
- Yun-Fei Dang
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Tian-Xiu Qiu
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Da-Wei Song
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China
| | - Lei Liu
- Laboratory of Biochemistry and Molecular Biology, School of Marine Sciences, Ningbo University, Ningbo, 315211, China; Key Laboratory of Applied Marine Biotechnology of Ministry of Education, Ningbo University, Ningbo, 315211, China.
| |
Collapse
|
8
|
Simó A, Cilleros-Mañé V, Just-Borràs L, Hurtado E, Nadal L, Tomàs M, Garcia N, Lanuza MA, Tomàs J. nPKCε Mediates SNAP-25 Phosphorylation of Ser-187 in Basal Conditions and After Synaptic Activity at the Neuromuscular Junction. Mol Neurobiol 2019; 56:5346-5364. [PMID: 30607888 DOI: 10.1007/s12035-018-1462-5] [Citation(s) in RCA: 12] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/05/2018] [Accepted: 12/18/2018] [Indexed: 12/14/2022]
Abstract
Protein kinase C (PKC) and substrates like SNAP-25 regulate neurotransmission. At the neuromuscular junction (NMJ), PKC promotes neurotransmitter release during synaptic activity. Thirty minutes of muscle contraction enhances presynaptic PKC isoform levels, specifically cPKCβI and nPKCε, through retrograde BDNF/TrkB signaling. This establishes a larger pool of these PKC isoforms ready to promote neuromuscular transmission. The PKC phosphorylation site in SNAP-25 has been mapped to the serine 187 (Ser-187), which is known to enhance calcium-dependent neurotransmitter release in vitro. Here, we localize SNAP-25 at the NMJ and investigate whether cPKCβI and/or nPKCε regulate SNAP-25 phosphorylation. We also investigate whether nerve and muscle cell activities regulate differently SNAP-25 phosphorylation and the involvement of BDNF/TrkB signaling. Our results demonstrate that nPKCε isoform is essential to positively regulate SNAP-25 phosphorylation on Ser-187 and that muscle contraction prevents it. TrkB and cPKCβI do not regulate SNAP-25 protein level or its phosphorylation during neuromuscular activity. The results provide evidence that nerve terminals need both pre- and postsynaptic activities to modulate SNAP-25 phosphorylation and ensure an accurate neurotransmission process.
Collapse
Affiliation(s)
- Anna Simó
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Victor Cilleros-Mañé
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Laia Just-Borràs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Erica Hurtado
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Laura Nadal
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Marta Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Neus Garcia
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Maria A Lanuza
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| | - Josep Tomàs
- Unitat d'Histologia i Neurobiologia (UHNEUROB), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| |
Collapse
|
9
|
Nowak G, Bakajsova-Takacsova D. Protein kinase Cε targets respiratory chain and mitochondrial membrane potential but not F 0 F 1 -ATPase in renal cells injured by oxidant. J Cell Biochem 2018; 119:9394-9407. [PMID: 30074270 PMCID: PMC6298597 DOI: 10.1002/jcb.27256] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/13/2018] [Accepted: 06/21/2018] [Indexed: 01/12/2023]
Abstract
We have previously shown that protein kinase Cε (PKCε) is involved in mitochondrial dysfunction in renal proximal tubular cells (RPTC). This study examined mitochondrial targets of active PKCε in RPTC injured by the model oxidant tert-butyl hydroperoxide (TBHP). TBHP exposure augmented the levels of phosphorylated (active) PKCε in mitochondria, which suggested translocation of PKCε to mitochondria after oxidant exposure. Oxidant injury decreased state 3 respiration, adenosine triphosphate (ATP) production, ATP content, and complex I activity. Further, TBHP exposure increased ΔΨm and production of reactive oxygen species (ROS), and induced mitochondrial fragmentation and RPTC death. PKCε activation by overexpressing constitutively active PKCε exacerbated decreases in state 3 respiration, complex I activity, ATP content, and augmented RPTC death. In contrast, inhibition of PKCε by overexpressing dnPKCε mutant restored state 3 respiration, respiratory control ratio, complex I activity, ΔΨm , and ATP production and content, but did not prevent decreases in F0 F1 -ATPase activity. Inhibition of PKCε prevented oxidant-induced production of ROS and mitochondrial fragmentation, and reduced RPTC death. We conclude that activation of PKCε mediates: (a) oxidant-induced changes in ΔΨm , decreases in mitochondrial respiration, complex I activity, and ATP content; (b) mitochondrial fragmentation; and (c) RPTC death. In contrast, oxidant-induced inhibition of F0 F1 -ATPase activity is not mediated by PKCε. These results show that, in contrast to the protective effects of PKCε in the heart, PKCε activation is detrimental to mitochondrial function and viability in RPTC and mediates oxidant-induced injury.
Collapse
Affiliation(s)
- Grazyna Nowak
- University of Arkansas for Medical Sciences, College of Pharmacy,
Department of Pharmaceutical Sciences, 4301 West Markham St., Little Rock, AR
72205
| | - Diana Bakajsova-Takacsova
- University of Arkansas for Medical Sciences, College of Pharmacy,
Department of Pharmaceutical Sciences, 4301 West Markham St., Little Rock, AR
72205
| |
Collapse
|
10
|
Nowak G, Takacsova-Bakajsova D, Megyesi J. Deletion of protein kinase C-ε attenuates mitochondrial dysfunction and ameliorates ischemic renal injury. Am J Physiol Renal Physiol 2016; 312:F109-F120. [PMID: 27760765 DOI: 10.1152/ajprenal.00115.2016] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2016] [Accepted: 10/11/2016] [Indexed: 02/08/2023] Open
Abstract
Previously, we documented that activation of protein kinase C-ε (PKC-ε) mediates mitochondrial dysfunction in cultured renal proximal tubule cells (RPTC). This study tested whether deletion of PKC-ε decreases dysfunction of renal cortical mitochondria and improves kidney function after renal ischemia. PKC-ε levels in mitochondria of ischemic kidneys increased 24 h after ischemia. Complex I- and complex II-coupled state 3 respirations were reduced 44 and 27%, respectively, in wild-type (WT) but unchanged and increased in PKC-ε-deficient (KO) mice after ischemia. Respiratory control ratio coupled to glutamate/malate oxidation decreased 50% in WT but not in KO mice. Activities of complexes I, III, and IV were decreased 59, 89, and 61%, respectively, in WT but not in KO ischemic kidneys. Proteomics revealed increases in levels of ATP synthase (α-subunit), complexes I and III, cytochrome oxidase, α-ketoglutarate dehydrogenase, and thioredoxin-dependent peroxide reductase after ischemia in KO but not in WT animals. PKC-ε deletion prevented ischemia-induced increases in oxidant production. Plasma creatinine levels increased 12-fold in WT and 3-fold in KO ischemic mice. PKC-ε deletion reduced tubular necrosis, brush border loss, and distal segment damage in ischemic kidneys. PKC-ε activation in hypoxic RPTC in primary culture exacerbated, whereas PKC-ε inhibition reduced, decreases in: 1) complex I- and complex II-coupled state 3 respirations and 2) activities of complexes I, III, and IV. We conclude that PKC-ε activation mediates 1) dysfunction of complexes I and III of the respiratory chain, 2) oxidant production, 3) morphological damage to the kidney, and 4) decreases in renal functions after ischemia.
Collapse
Affiliation(s)
- Grazyna Nowak
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas; and
| | - Diana Takacsova-Bakajsova
- Department of Pharmaceutical Sciences, College of Pharmacy, University of Arkansas for Medical Sciences, Little Rock, Arkansas; and
| | - Judit Megyesi
- Division of Nephrology, Department of Physiology and Biophysics, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| |
Collapse
|
11
|
Capuani B, Pacifici F, Pastore D, Palmirotta R, Donadel G, Arriga R, Bellia A, Di Daniele N, Rogliani P, Abete P, Sbraccia P, Guadagni F, Lauro D, Della-Morte D. The role of epsilon PKC in acute and chronic diseases: Possible pharmacological implications of its modulators. Pharmacol Res 2016; 111:659-667. [PMID: 27461137 DOI: 10.1016/j.phrs.2016.07.029] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/22/2016] [Accepted: 07/22/2016] [Indexed: 02/06/2023]
Abstract
Epsilon Protein kinase C (εPCK) is a particular kinase that, when activated, is able to protect against different stress injuries and therefore has been proposed to be a potential molecular target against acute and chronic diseases. Particular attention has been focused on εPCK for its involvement in the protective mechanism of Ischemic Preconditioning (IPC), a powerful endogenous mechanism characterized by subthreshold ischemic insults able to protect organs against ischemic injury. Therefore, in the past decades several εPCK modulators have been tested with the object to emulate εPCK mediate protection. Among these the most promising, so far, has been the ΨεRACK peptide, a homologous of RACK receptor for εPKC, that when administrated can mimic its effect in the cells. However, results from studies on εPCK indicate controversial role of this kinase in different organs and diseases, such as myocardial infarct, stroke, diabetes and cancer. Therefore, in this review we provide a discussion on the function of εPCK in acute and chronic diseases and how the different activators and inhibitors have been used to modulate its activity. A better understanding of its function is still needed to definitively target εPCK as novel therapeutic strategy.
Collapse
Affiliation(s)
- Barbara Capuani
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Francesca Pacifici
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Donatella Pastore
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Raffaele Palmirotta
- Department of Biomedical Sciences and Human Oncology, University of Bari Aldo Moro, Bari, Italy
| | - Giulia Donadel
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Roberto Arriga
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Alfonso Bellia
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Nicola Di Daniele
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Paola Rogliani
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Pasquale Abete
- Department of Translational Medical Sciences, University of Naples Federico II, Naples, Italy
| | - Paolo Sbraccia
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - Fiorella Guadagni
- IRCCS San Raffaele Pisana, Rome, Italy; San Raffaele Roma Open University, Via di Val Cannuta 247, 00166 Rome, Italy
| | - Davide Lauro
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy
| | - David Della-Morte
- Department of Systems Medicine, School of Medicine, University of Rome Tor Vergata, Rome, Italy; IRCCS San Raffaele Pisana, Rome, Italy.
| |
Collapse
|
12
|
Vyas FS, Hargreaves AJ, Bonner PL, Boocock DJ, Coveney C, Dickenson JM. A1 adenosine receptor-induced phosphorylation and modulation of transglutaminase 2 activity in H9c2 cells: A role in cell survival. Biochem Pharmacol 2016; 107:41-58. [DOI: 10.1016/j.bcp.2016.03.016] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2016] [Accepted: 03/17/2016] [Indexed: 12/25/2022]
|
13
|
Zhao EY, Efendizade A, Cai L, Ding Y. The role of Akt (protein kinase B) and protein kinase C in ischemia-reperfusion injury. Neurol Res 2016; 38:301-8. [PMID: 27092987 DOI: 10.1080/01616412.2015.1133024] [Citation(s) in RCA: 43] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023]
Abstract
Stroke is a leading cause of long-term disability and death in the United States. Currently, tissue plasminogen activator (tPA), is the only Food and Drug Administration-approved treatment for acute ischemic stroke. However, the use of tPA is restricted to a small subset of acute stroke patients due to its limited 3-h therapeutic time window. Given the limited therapeutic options at present and the multi-factorial progression of ischemic stroke, emphasis has been placed on the discovery and use of combination therapies aimed at various molecular targets contributing to ischemic cell death. Protein kinase C (PKC) and Akt (protein kinase B) are serine/threonine kinases that play a critical role in mediating ischemic-reperfusion injury and cellular growth and survival, respectively. The present review will examine the role of PKC and Akt in the cellular response to ischemic-reperfusion injury.
Collapse
Affiliation(s)
- Ethan Y Zhao
- a Departmentof Neurosurgery , Wayne State University School of Medicine , Detroit , MI 48201 , USA
| | - Aslan Efendizade
- b Michigan State University College of Osteopathic Medicine , East Lansing , MI 48825 , USA
| | - Lipeng Cai
- c Department of Neurology , China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University , Beijing , China
| | - Yuchuan Ding
- a Departmentof Neurosurgery , Wayne State University School of Medicine , Detroit , MI 48201 , USA.,c Department of Neurology , China-America Institute of Neuroscience, Luhe Hospital, Capital Medical University , Beijing , China
| |
Collapse
|
14
|
Guo C, Wang S, Duan J, Jia N, Zhu Y, Ding Y, Guan Y, Wei G, Yin Y, Xi M, Wen A. Protocatechualdehyde Protects Against Cerebral Ischemia-Reperfusion-Induced Oxidative Injury Via Protein Kinase Cε/Nrf2/HO-1 Pathway. Mol Neurobiol 2016; 54:833-845. [PMID: 26780453 DOI: 10.1007/s12035-016-9690-z] [Citation(s) in RCA: 84] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2015] [Accepted: 01/05/2016] [Indexed: 11/24/2022]
Abstract
Oxidative stress is closely related to the pathogenesis of ischemic stroke. Protocatechualdehyde (PCA) is a phenolic acid compound that has the putative antioxidant activities. The present study was aimed to investigate the molecular mechanisms involved in the antioxidative effect of PCA against cerebral ischemia/reperfusion (I/R) injury. The experiment stroke model was produced in Sprague-Dawley rats via middle cerebral artery occlusion (MCAO). To model ischemia-like conditions in vitro, differentiated SH-SY5Y cells were exposed to transient oxygen and glucose deprivation (OGD). Treatment with PCA significantly improved neurologic score, reduced infarct volume and necrotic neurons, and also decreased reactive oxygen species (ROS) production, 4-hydroxynonenal (4-HNE), and 8-hydroxy-2'-deoxyguanosine (8-OHdG) contents at 24 h after reperfusion. Meanwhile, PCA significantly increased the transcription nuclear factor erythroid 2-related factor 2 (Nrf2) and heme oxygenase-1 (HO-1) expressions in the ischemic cerebral cortex as shown by immunofluorescence staining and Western blot analysis. In vitro experiment showed that PCA protected differentiated SH-SY5Y cells against OGD-induced injury. Likewise, PCA also increased markedly the Nrf2 and HO-1 expressions in a dose-dependent manner. The neuroprotection effect of PCA was abolished by knockdown of Nrf2 and HO-1. Moreover, knockdown of protein kinase Cε (PKCε) also blocked PCA-induced Nfr2 nuclear translocation, HO-1 expression, and neuroprotection. Taken together, these results provide evidences that PCA can protect against cerebral ischemia-reperfusion-induced oxidative injury, and the neuroprotective effect involves the PKCε/Nrf2/HO-1 pathway.
Collapse
Affiliation(s)
- Chao Guo
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Shiquan Wang
- Department of Anesthesiology, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Jialin Duan
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Na Jia
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Yanrong Zhu
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Yi Ding
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Yue Guan
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Guo Wei
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Ying Yin
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China
| | - Miaomaio Xi
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China.
| | - Aidong Wen
- Department of Pharmacy, Xijing Hospital, Fourth Military Medical University, Xi'an, 710032, People's Republic of China.
| |
Collapse
|
15
|
Obis T, Hurtado E, Nadal L, Tomàs M, Priego M, Simon A, Garcia N, Santafe MM, Lanuza MA, Tomàs J. The novel protein kinase C epsilon isoform modulates acetylcholine release in the rat neuromuscular junction. Mol Brain 2015; 8:80. [PMID: 26625935 PMCID: PMC4665914 DOI: 10.1186/s13041-015-0171-5] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2015] [Accepted: 11/25/2015] [Indexed: 01/20/2023] Open
Abstract
Background Various protein kinase C (PKC) isoforms contribute to the phosphorylating activity that modulates neurotransmitter release. In previous studies we showed that nPKCε is confined in the presynaptic site of the neuromuscular junction and its presynaptic function is activity-dependent. Furthermore, nPKCε regulates phorbol ester-induced acetylcholine release potentiation, which further indicates that nPKCε is involved in neurotransmission. The present study is designed to examine the nPKCε involvement in transmitter release at the neuromuscular junction. Results We use the specific nPKCε translocation inhibitor peptide εV1-2 and electrophysiological experiments to investigate the involvement of this isoform in acetylcholine release. We observed that nPKCε membrane translocation is key to the synaptic potentiation of NMJ, being involved in several conditions that upregulate PKC isoforms coupling to acetylcholine (ACh) release (incubation with high Ca2+, stimulation with phorbol esters and protein kinase A, stimulation with adenosine 3′,5′-cyclic monophosphorothioate, 8-Bromo-, Rp-isomer, sodium salt -Sp-8-BrcAMP-). In all these conditions, preincubation with the nPKCε translocation inhibitor peptide (εV1-2) impairs PKC coupling to acetylcholine release potentiation. In addition, the inhibition of nPKCε translocation and therefore its activity impedes that presynaptic muscarinic autoreceptors and adenosine autoreceptors modulate transmitter secretion. Conclusions Together, these results point to the importance of nPKCε isoform in the control of acetylcholine release in the neuromuscular junction.
Collapse
Affiliation(s)
- Teresa Obis
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Erica Hurtado
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Laura Nadal
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Marta Tomàs
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Mercedes Priego
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Anna Simon
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Neus Garcia
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Manel M Santafe
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain
| | - Maria A Lanuza
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| | - Josep Tomàs
- Unitat d'Histologia i Neurobiologia (UHN), Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| |
Collapse
|
16
|
Ford A, Castonguay A, Cottet M, Little JW, Chen Z, Symons-Liguori AM, Doyle T, Egan TM, Vanderah TW, De Koninck Y, Tosh DK, Jacobson KA, Salvemini D. Engagement of the GABA to KCC2 signaling pathway contributes to the analgesic effects of A3AR agonists in neuropathic pain. J Neurosci 2015; 35:6057-67. [PMID: 25878279 PMCID: PMC4397603 DOI: 10.1523/jneurosci.4495-14.2015] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2014] [Revised: 02/27/2015] [Accepted: 03/05/2015] [Indexed: 01/18/2023] Open
Abstract
More than 1.5 billion people worldwide suffer from chronic pain, yet current treatment strategies often lack efficacy or have deleterious side effects in patients. Adenosine is an inhibitory neuromodulator that was previously thought to mediate antinociception through the A1 and A2A receptor subtypes. We have since demonstrated that A3AR agonists have potent analgesic actions in preclinical rodent models of neuropathic pain and that A3AR analgesia is independent of adenosine A1 or A2A unwanted effects. Herein, we explored the contribution of the GABA inhibitory system to A3AR-mediated analgesia using well-characterized mouse and rat models of chronic constriction injury (CCI)-induced neuropathic pain. The deregulation of GABA signaling in pathophysiological pain states is well established: GABA signaling can be hampered by a reduction in extracellular GABA synthesis by GAD65 and enhanced extracellular GABA reuptake via the GABA transporter, GAT-1. In neuropathic pain, GABAAR-mediated signaling can be further disrupted by the loss of the KCC2 chloride anion gradient. Here, we demonstrate that A3AR agonists (IB-MECA and MRS5698) reverse neuropathic pain via a spinal mechanism of action that modulates GABA activity. Spinal administration of the GABAA antagonist, bicuculline, disrupted A3AR-mediated analgesia. Furthermore, A3AR-mediated analgesia was associated with reductions in CCI-related GAD65 and GAT-1 serine dephosphorylation as well as an enhancement of KCC2 serine phosphorylation and activity. Our results suggest that A3AR-mediated reversal of neuropathic pain increases modulation of GABA inhibitory neurotransmission both directly and indirectly through protection of KCC2 function, underscoring the unique utility of A3AR agonists in chronic pain.
Collapse
Affiliation(s)
- Amanda Ford
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Annie Castonguay
- Institut Universitaire en Santé Mentale de Québec, Québec City, Quebec G1J 2G3, Canada, Department of Psychiatry & Neuroscience, Université Laval, Québec City, Quebec G1K 7P4, Canada
| | - Martin Cottet
- Institut Universitaire en Santé Mentale de Québec, Québec City, Quebec G1J 2G3, Canada, Department of Psychiatry & Neuroscience, Université Laval, Québec City, Quebec G1K 7P4, Canada
| | - Joshua W Little
- Department of Surgery, Center for Anatomical Science and Education, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Zhoumou Chen
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Ashley M Symons-Liguori
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724-5050, and
| | - Timothy Doyle
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Terrance M Egan
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104
| | - Todd W Vanderah
- Department of Pharmacology, College of Medicine, University of Arizona, Tucson, Arizona 85724-5050, and
| | - Yves De Koninck
- Institut Universitaire en Santé Mentale de Québec, Québec City, Quebec G1J 2G3, Canada, Department of Psychiatry & Neuroscience, Université Laval, Québec City, Quebec G1K 7P4, Canada
| | - Dilip K Tosh
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0810
| | - Kenneth A Jacobson
- Molecular Recognition Section, Laboratory of Bioorganic Chemistry, National Institute of Diabetes and Digestive and Kidney Diseases, National Institutes of Health, Bethesda, Maryland 20892-0810
| | - Daniela Salvemini
- Department of Pharmacological and Physiological Science, St. Louis University School of Medicine, St. Louis, Missouri 63104,
| |
Collapse
|
17
|
The divergent roles of autophagy in ischemia and preconditioning. Acta Pharmacol Sin 2015; 36:411-20. [PMID: 25832421 PMCID: PMC4387298 DOI: 10.1038/aps.2014.151] [Citation(s) in RCA: 59] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/03/2014] [Accepted: 12/20/2014] [Indexed: 12/11/2022]
Abstract
Autophagy is an evolutionarily conserved and lysosome-dependent process for degrading and recycling cellular constituents. Autophagy is activated following an ischemic insult or preconditioning, but it may exert dual roles in cell death or survival during these two processes. Preconditioning or lethal ischemia may trigger autophagy via multiple signaling pathways involving endoplasmic reticulum (ER) stress, AMPK/TSC/mTOR, Beclin 1/BNIP3/SPK2, and FoxO/NF-κB transcription factors, etc. Autophagy then interacts with apoptotic and necrotic signaling pathways to regulate cell death. Autophagy may also maintain cell function by removing protein aggregates or damaged mitochondria. To date, the dual roles of autophagy in ischemia and preconditioning have not been fully clarified. The purpose of the present review is to summarize the recent progress in the mechanisms underlying autophagy activation during ischemia and preconditioning. A better understanding of the dual effects of autophagy in ischemia and preconditioning could help to develop new strategies for the preventive treatment of ischemia.
Collapse
|
18
|
Obis T, Besalduch N, Hurtado E, Nadal L, Santafe MM, Garcia N, Tomàs M, Priego M, Lanuza MA, Tomàs J. The novel protein kinase C epsilon isoform at the adult neuromuscular synapse: location, regulation by synaptic activity-dependent muscle contraction through TrkB signaling and coupling to ACh release. Mol Brain 2015; 8:8. [PMID: 25761522 PMCID: PMC4348107 DOI: 10.1186/s13041-015-0098-x] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/10/2014] [Accepted: 01/16/2015] [Indexed: 12/03/2022] Open
Abstract
Background Protein kinase C (PKC) regulates a variety of neural functions, including neurotransmitter release. Although various PKC isoforms can be expressed at the synaptic sites and specific cell distribution may contribute to their functional diversity, little is known about the isoform-specific functions of PKCs in neuromuscular synapse. The present study is designed to examine the location of the novel isoform nPKCε at the neuromuscular junction (NMJ), their synaptic activity-related expression changes, its regulation by muscle contraction, and their possible involvement in acetylcholine release. Results We use immunohistochemistry and confocal microscopy to demonstrate that the novel isoform nPKCε is exclusively located in the motor nerve terminals of the adult rat NMJ. We also report that electrical stimulation of synaptic inputs to the skeletal muscle significantly increased the amount of nPKCε isoform as well as its phosphorylated form in the synaptic membrane, and muscle contraction is necessary for these nPKCε expression changes. The results also demonstrate that synaptic activity-induced muscle contraction promotes changes in presynaptic nPKCε through the brain-derived neurotrophic factor (BDNF)-mediated tyrosine kinase receptor B (TrkB) signaling. Moreover, nPKCε activity results in phosphorylation of the substrate MARCKS involved in actin cytoskeleton remodeling and related with neurotransmission. Finally, blocking nPKCε with a nPKCε-specific translocation inhibitor peptide (εV1-2) strongly reduces phorbol ester-induced ACh release potentiation, which further indicates that nPKCε is involved in neurotransmission. Conclusions Together, these results provide a mechanistic insight into how synaptic activity-induced muscle contraction could regulate the presynaptic action of the nPKCε isoform and suggest that muscle contraction is an important regulatory step in TrkB signaling at the NMJ.
Collapse
Affiliation(s)
- Teresa Obis
- Unitat d'Histologia i Neurobiologia (UHN). Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| | - Núria Besalduch
- Unitat d'Histologia i Neurobiologia (UHN). Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| | - Erica Hurtado
- Unitat d'Histologia i Neurobiologia (UHN). Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| | - Laura Nadal
- Unitat d'Histologia i Neurobiologia (UHN). Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| | - Manel M Santafe
- Unitat d'Histologia i Neurobiologia (UHN). Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| | - Neus Garcia
- Unitat d'Histologia i Neurobiologia (UHN). Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| | - Marta Tomàs
- Unitat d'Histologia i Neurobiologia (UHN). Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| | - Mercedes Priego
- Unitat d'Histologia i Neurobiologia (UHN). Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| | - Maria A Lanuza
- Unitat d'Histologia i Neurobiologia (UHN). Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| | - Josep Tomàs
- Unitat d'Histologia i Neurobiologia (UHN). Facultat de Medicina i Ciències de la Salut, Universitat Rovira i Virgili, Sant Llorenç 21, 43201, Reus, Spain.
| |
Collapse
|
19
|
The "memory kinases": roles of PKC isoforms in signal processing and memory formation. PROGRESS IN MOLECULAR BIOLOGY AND TRANSLATIONAL SCIENCE 2014; 122:31-59. [PMID: 24484697 DOI: 10.1016/b978-0-12-420170-5.00002-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
The protein kinase C (PKC) isoforms, which play an essential role in transmembrane signal conduction, can be viewed as a family of "memory kinases." Evidence is emerging that they are critically involved in memory acquisition and maintenance, in addition to their involvement in other functions of cells. Deficits in PKC signal cascades in neurons are one of the earliest abnormalities in the brains of patients suffering from Alzheimer's disease. Their dysfunction is also involved in several other types of memory impairments, including those related to emotion, mental retardation, brain injury, and vascular dementia/ischemic stroke. Inhibition of PKC activity leads to a reduced capacity of many types of learning and memory, but may have therapeutic values in treating substance abuse or aversive memories. PKC activators, on the other hand, have been shown to possess memory-enhancing and antidementia actions. PKC pharmacology may, therefore, represent an attractive area for developing effective cognitive drugs for the treatment of many types of memory disorders and dementias.
Collapse
|
20
|
Ferrari LF, Levine E, Levine JD. Role of a novel nociceptor autocrine mechanism in chronic pain. Eur J Neurosci 2013; 37:1705-13. [PMID: 23379641 DOI: 10.1111/ejn.12145] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2012] [Revised: 12/11/2012] [Accepted: 01/07/2013] [Indexed: 12/13/2022]
Abstract
We have previously shown, in the rat, that neuropathic and inflammatory events produce a neuroplastic change in nociceptor function whereby a subsequent exposure to a proinflammatory mediator (e.g. prostaglandin E2 ; PGE2 ) produces markedly prolonged mechanical hyperalgesia. While the initial approximately 30 min of this prolonged PGE2 hyperalgesia remains PKA-dependent, it subsequently switches to become dependent on protein kinase C epsilon (PKCε). In this study we tested the hypothesis that the delayed onset, PKCε-mediated, component of PGE2 hyperalgesia is generated by the active release of a nucleotide from the peripheral terminal of the primed nociceptor and this nucleotide is then metabolized to produce adenosine, which acts on a Gi-coupled A1 adenosine receptor on the nociceptor to generate PKCε-dependent hyperalgesia. We report that inhibitors of ATP-binding cassette transporters, of ecto-5'-phosphodiesterase and ecto-5'nucleotidase (enzymes involved in the metabolism of cyclic nucleotides to adenosine) and of A1 adenosine receptors each eliminated the late, but not the early, phase of PGE2 -induced hyperalgesia in primed animals. A second model of chronic pain induced by transient attenuation of G-protein-coupled receptor kinase 2, in which the prolongation of PGE2 hyperalgesia is not PKCε-dependent, was not attenuated by inhibitors of any of these mechanisms. Based on these results we propose a contribution of an autocrine mechanism, in the peripheral terminal of the nociceptor, in the hyperalgesic priming model of chronic pain.
Collapse
Affiliation(s)
- Luiz F Ferrari
- Division of Neuroscience, Departments of Medicine and Oral Surgery, University of California, San Francisco, CA 94143-0440, USA
| | | | | |
Collapse
|
21
|
Zhao H, Ren C, Chen X, Shen J. From rapid to delayed and remote postconditioning: the evolving concept of ischemic postconditioning in brain ischemia. Curr Drug Targets 2012; 13:173-87. [PMID: 22204317 DOI: 10.2174/138945012799201621] [Citation(s) in RCA: 89] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/01/2010] [Revised: 08/09/2011] [Accepted: 08/12/2011] [Indexed: 01/13/2023]
Abstract
Ischemic postconditioning is a concept originally defined to contrast with that of ischemic preconditioning. While both preconditioning and postconditioning confer a neuroprotective effect on brain ischemia, preconditioning is a sublethal insult performed in advance of brain ischemia, and postconditioning, which conventionally refers to a series of brief occlusions and reperfusions of the blood vessels, is conducted after ischemia/reperfusion. In this article, we first briefly review the history of preconditioning, including the experimentation that initially uncovered its neuroprotective effects and later revealed its underlying mechanisms-of-action. We then discuss how preconditioning research evolved into that of postconditioning--a concept that now represents a broad range of stimuli or triggers, including delayed postconditioning, pharmacological postconditioning, remote postconditioning--and its underlying protective mechanisms involving the Akt, MAPK, PKC and K(ATP) channel cell-signaling pathways. Because the concept of postconditioning is so closely associated with that of preconditioning, and both share some common protective mechanisms, we also discuss whether a combination of preconditioning and postconditioning offers greater protection than preconditioning or postconditioning alone.
Collapse
Affiliation(s)
- Heng Zhao
- Department of Neurosurgery, Stanford University School of Medicine, Stanford, CA 94305-5327, USA.
| | | | | | | |
Collapse
|
22
|
Dave KR, Christian SL, Perez-Pinzon MA, Drew KL. Neuroprotection: lessons from hibernators. Comp Biochem Physiol B Biochem Mol Biol 2012; 162:1-9. [PMID: 22326449 PMCID: PMC3334476 DOI: 10.1016/j.cbpb.2012.01.008] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Revised: 01/26/2012] [Accepted: 01/30/2012] [Indexed: 12/15/2022]
Abstract
Mammals that hibernate experience extreme metabolic states and body temperatures as they transition between euthermia, a state resembling typical warm blooded mammals, and prolonged torpor, a state of suspended animation where the brain receives as low as 10% of normal cerebral blood flow. Transitions into and out of torpor are more physiologically challenging than the extreme metabolic suppression and cold body temperatures of torpor per se. Mammals that hibernate show unprecedented capacities to tolerate cerebral ischemia, a decrease in blood flow to the brain caused by stroke, cardiac arrest or brain trauma. While cerebral ischemia often leads to death or disability in humans and most other mammals, hibernating mammals suffer no ill effects when blood flow to the brain is dramatically decreased during torpor or experimentally induced during euthermia. These animals, as adults, also display rapid and pronounced synaptic flexibility where synapses retract during torpor and rapidly re-emerge upon arousal. A variety of coordinated adaptations contribute to tolerance of cerebral ischemia in these animals. In this review we discuss adaptations in heterothermic mammals that may suggest novel therapeutic targets and strategies to protect the human brain against cerebral ischemic damage and neurodegenerative disease.
Collapse
Affiliation(s)
- Kunjan R Dave
- Cerebral Vascular Disease Research Laboratories, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, FL 33136, USA.
| | | | | | | |
Collapse
|
23
|
Sun MK, Alkon DL. Activation of protein kinase C isozymes for the treatment of dementias. ADVANCES IN PHARMACOLOGY (SAN DIEGO, CALIF.) 2012; 64:273-302. [PMID: 22840750 DOI: 10.1016/b978-0-12-394816-8.00008-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Memories are much more easily impaired than improved. Dementias, a lasting impairment of memory function, occur in a variety of cognitive disorders and become more clinically dominant as the population ages. Protein kinase C is one of the "cognitive kinases," and plays an essential role in both memory acquisition and maintenance. Deficits in protein kinase C (PKC) signal cascades in neurons represent one of the earliest changes in the brains of patients with Alzheimer's disease (AD) and other types of memory impairment, including those related to cerebral ischemia and ischemic stroke. Inhibition or impairment of PKC activity results in compromised learning and memory, whereas an appropriate activation of certain PKC isozymes leads to an enhancement of learning and memory and/or antidementic effects. In preclinical studies, PKC activators have been shown to increase the expression and activity of PKC isozymes, thereby restoring PKC signaling and downstream activity, including stimulation of neurotrophic activity, synaptic/structural remodeling, and synaptogenesis in the hippocampus and related cortical areas. PKC activators also reduce the accumulation of neurotoxic amyloid and tau protein hyperphosphorylation and support anti-apoptotic processes in the brain. These observations strongly suggest that PKC pharmacology may represent an attractive area for the development of effective cognition-enhancing therapeutics for the treatment of dementias.
Collapse
Affiliation(s)
- Miao-Kun Sun
- Blanchette Rockefeller Neurosciences Institute, Morgantown, WV, USA
| | | |
Collapse
|
24
|
Nowak G, Bakajsova D, Samarel AM. Protein kinase C-epsilon activation induces mitochondrial dysfunction and fragmentation in renal proximal tubules. Am J Physiol Renal Physiol 2011; 301:F197-208. [PMID: 21289057 DOI: 10.1152/ajprenal.00364.2010] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
PKC-ε activation mediates protection from ischemia-reperfusion injury in the myocardium. Mitochondria are a subcellular target of these protective mechanisms of PKC-ε. Previously, we have shown that PKC-ε activation is involved in mitochondrial dysfunction in oxidant-injured renal proximal tubular cells (RPTC; Nowak G, Bakajsova D, Clifton GL Am J Physiol Renal Physiol 286: F307-F316, 2004). The goal of this study was to examine the role of PKC-ε activation in mitochondrial dysfunction and to identify mitochondrial targets of PKC-ε in RPTC. The constitutively active and inactive mutants of PKC-ε were overexpressed in primary cultures of RPTC using the adenoviral technique. Increases in active PKC-ε levels were accompanied by PKC-ε translocation to mitochondria. Sustained PKC-ε activation resulted in decreases in state 3 respiration, electron transport rate, ATP production, ATP content, and activities of complexes I and IV and F(0)F(1)-ATPase. Furthermore, PKC-ε activation increased mitochondrial membrane potential and oxidant production and induced mitochondrial fragmentation and RPTC death. Accumulation of the dynamin-related protein in mitochondria preceded mitochondrial fragmentation. Antioxidants blocked PKC-ε-induced increases in the oxidant production but did not prevent mitochondrial fragmentation and cell death. The inactive PKC-ε mutant had no effect on mitochondrial functions, morphology, oxidant production, and RPTC viability. We conclude that active PKC-ε targets complexes I and IV and F(0)F(1)-ATPase in RPTC. PKC-ε activation mediates mitochondrial dysfunction, hyperpolarization, and fragmentation. It also induces oxidant generation and cell death, but oxidative stress is not the mechanism of RPTC death. These results show that in contrast to protective effects of PKC-ε activation in cardiomyocytes, sustained PKC-ε activation is detrimental to mitochondrial function and viability in RPTC.
Collapse
Affiliation(s)
- Grazyna Nowak
- University of Arkansas for Medical Sciences, Dept. of Pharmaceutical Sciences, 4301 West Markham St., Little Rock, AR 72205, USA.
| | | | | |
Collapse
|
25
|
Wang Q, Li X, Chen Y, Wang F, Yang Q, Chen S, Min Y, Li X, Xiong L. Activation of Epsilon Protein Kinase C-Mediated Anti-Apoptosis Is Involved in Rapid Tolerance Induced by Electroacupuncture Pretreatment Through Cannabinoid Receptor Type 1. Stroke 2011; 42:389-96. [PMID: 21183751 DOI: 10.1161/strokeaha.110.597336] [Citation(s) in RCA: 81] [Impact Index Per Article: 5.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Affiliation(s)
- Qiang Wang
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Xuying Li
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Yanke Chen
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Feng Wang
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Qianzi Yang
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Shaoyang Chen
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Yuyuan Min
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Xin Li
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| | - Lize Xiong
- From the Department of Anesthesiology (Q.W., X.L., F.W., Q.Y., S.Y., Y.M., L.X., L.X.), Xijing Hospital, Fourth Military Medical University, Xi'an, China; Center for Biomedical Research on Pain (Y.C.), College of Medicine, Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
26
|
Protein kinase Cμ mediates adenosine-stimulated steroidogenesis in primary rat adrenal cells. FEBS Lett 2010; 584:4442-8. [DOI: 10.1016/j.febslet.2010.10.001] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2010] [Revised: 09/17/2010] [Accepted: 10/06/2010] [Indexed: 11/22/2022]
|
27
|
Dave KR, Anthony Defazio R, Raval AP, Dashkin O, Saul I, Iceman KE, Perez-Pinzon MA, Drew KL. Protein kinase C epsilon activation delays neuronal depolarization during cardiac arrest in the euthermic arctic ground squirrel. J Neurochem 2009; 110:1170-9. [PMID: 19493168 PMCID: PMC2774829 DOI: 10.1111/j.1471-4159.2009.06196.x] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
During the pre-hibernation season, arctic ground squirrels (AGS) can tolerate 8 min of asphyxial cardiac arrest (CA) without detectable brain pathology. Better understanding of the mechanisms regulating innate ischemia tolerance in AGS has the potential to facilitate the development of novel prophylactic agents to induce ischemic tolerance in patients at risk of stroke or CA. We hypothesized that neuroprotection in AGS involves robust maintenance of ion homeostasis similar to anoxia-tolerant turtles. Ion homeostasis was assessed by monitoring ischemic depolarization (ID) in cerebral cortex during CA in vivo and during oxygen glucose deprivation in vitro in acutely prepared hippocampal slices. In both models, the onset of ID was significantly delayed in AGS compared with rats. The epsilon protein kinase C (epsilonPKC) is a key mediator of neuroprotection and inhibits both Na+/K+-ATPase and voltage-gated sodium channels, primary mediators of the collapse of ion homeostasis during ischemia. The selective peptide inhibitor of epsilonPKC (epsilonV1-2) shortened the time to ID in brain slices from AGS but not in rats despite evidence that epsilonV1-2 decreased activation of epsilonPKC in brain slices from both rats and AGS. These results support the hypothesis that epsilonPKC activation delays the collapse of ion homeostasis during ischemia in AGS.
Collapse
Affiliation(s)
- Kunjan R Dave
- Department of Neurology, Cerebral Vascular Disease Research Center, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida 33101, USA.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Abstract
Brain ischemia is one of the most common causes of death and the leading cause of adult disability in the world. Brain ischemic preconditioning (BIP) refers to a transient, sublethal ischemia which results in tolerance to later, otherwise lethal, cerebral ischemia. Many attempts have been made to understand the molecular and cellular mechanisms underlying the neuroprotection offered by ischemic preconditioning. Many studies have shown that neuroprotective mechanisms may involve a series of molecular regulatory pathways including activation of the N-methyl-D-aspartate (NMDA) and adenosine receptors; activation of intracellular signaling pathways such as mitogen activated protein kinases (MAPK) and other protein kinases; upregulation of Bcl-2 and heat shock proteins (HSPs); and activation of the ubiquitin-proteasome pathway and the autophagic-lysosomal pathway. A better understanding of the processes that lead to cell death after stroke as well as of the endogenous neuroprotective mechanisms by which BIP protects against brain ischemic insults could help to develop new therapeutic strategies for this devastating neurological disease. The purpose of the present review is to summarize the neuroprotective mechanisms of BIP and to discuss the possibility of mimicking ischemic preconditioning as a new strategy for preventive treatment of ischemia.
Collapse
|
29
|
Muscella A, Urso L, Calabriso N, Vetrugno C, Fanizzi FP, Storelli C, Marsigliante S. Functions of epidermal growth factor receptor in cisplatin response of thyroid cells. Biochem Pharmacol 2009; 77:979-92. [DOI: 10.1016/j.bcp.2008.12.001] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2008] [Revised: 11/28/2008] [Accepted: 12/01/2008] [Indexed: 11/28/2022]
|
30
|
DeFazio RA, Raval AP, Lin HW, Dave KR, Della-Morte D, Perez-Pinzon MA. GABA synapses mediate neuroprotection after ischemic and epsilonPKC preconditioning in rat hippocampal slice cultures. J Cereb Blood Flow Metab 2009; 29:375-84. [PMID: 18957990 PMCID: PMC2696173 DOI: 10.1038/jcbfm.2008.126] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Delayed neuroprotection against ischemic challenges is conferred by both ischemic preconditioning (IPC) and preconditioning by activation of the epsilon-isoform of protein kinase C (epsilonPKC-PC). In vivo, ischemic preconditioning enhances GABA release and ameliorates glutamate release during lethal cerebral ischemia. We tested the hypothesis that IPC and epsilonPKC-PC confer neuroprotection by GABA synapses in rat organotypic hippocampal slices. Ischemic preconditioning or epsilonPKC-PC was induced with 15 mins oxygen-glucose deprivation (OGD) or psiepsilonRACK, a selective epsilonPKC activator; and test ischemia consisted of 40 mins OGD. At the time of peak neuroprotection (48 h after preconditioning), we recorded GABA(A) receptor-mediated miniature postsynaptic currents (GABA mPSCs) in vulnerable CA1 pyramidal neurons using whole-cell voltage clamp techniques. The frequency and amplitude of GABA mPSCs significantly increased 48 h after IPC. In contrast, epsilonPKC-PC enhanced only the amplitude of GABA mPSCs with no effect on frequency. We next asked if neuroprotection depended on these changes in GABA synapses. Weak antagonism of the GABA(A) receptor with bicuculline (100 nmol/L) decreased the amplitude of GABA mPSCs by 20.9+/-6.1%. When applied during test ischemia, 100 nmol/L bicuculline abolished neuroprotection conferred by either IPC or epsilonPKC-PC. We conclude that neuroprotection conferred by preconditioning depends on functional modifications of GABA synapses.
Collapse
Affiliation(s)
- R Anthony DeFazio
- Cerebral Vascular Disease Research Center, Department of Neurology, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida 33101, USA.
| | | | | | | | | | | |
Collapse
|
31
|
Stone TW, Ceruti S, Abbracchio MP. Adenosine receptors and neurological disease: neuroprotection and neurodegeneration. Handb Exp Pharmacol 2009:535-87. [PMID: 19639293 DOI: 10.1007/978-3-540-89615-9_17] [Citation(s) in RCA: 146] [Impact Index Per Article: 9.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Adenosine receptors modulate neuronal and synaptic function in a range of ways that may make them relevant to the occurrence, development and treatment of brain ischemic damage and degenerative disorders. A(1) adenosine receptors tend to suppress neural activity by a predominantly presynaptic action, while A(2A) adenosine receptors are more likely to promote transmitter release and postsynaptic depolarization. A variety of interactions have also been described in which adenosine A(1) or A(2) adenosine receptors can modify cellular responses to conventional neurotransmitters or receptor agonists such as glutamate, NMDA, nitric oxide and P2 purine receptors. Part of the role of adenosine receptors seems to be in the regulation of inflammatory processes that often occur in the aftermath of a major insult or disease process. All of the adenosine receptors can modulate the release of cytokines such as interleukins and tumor necrosis factor-alpha from immune-competent leukocytes and glia. When examined directly as modifiers of brain damage, A(1) adenosine receptor (AR) agonists, A(2A)AR agonists and antagonists, as well as A(3)AR antagonists, can protect against a range of insults, both in vitro and in vivo. Intriguingly, acute and chronic treatments with these ligands can often produce diametrically opposite effects on damage outcome, probably resulting from adaptational changes in receptor number or properties. In some cases molecular approaches have identified the involvement of ERK and GSK-3beta pathways in the protection from damage. Much evidence argues for a role of adenosine receptors in neurological disease. Receptor densities are altered in patients with Alzheimer's disease, while many studies have demonstrated effects of adenosine and its antagonists on synaptic plasticity in vitro, or on learning adequacy in vivo. The combined effects of adenosine on neuronal viability and inflammatory processes have also led to considerations of their roles in Lesch-Nyhan syndrome, Creutzfeldt-Jakob disease, Huntington's disease and multiple sclerosis, as well as the brain damage associated with stroke. In addition to the potential pathological relevance of adenosine receptors, there are earnest attempts in progress to generate ligands that will target adenosine receptors as therapeutic agents to treat some of these disorders.
Collapse
Affiliation(s)
- Trevor W Stone
- Institute of Biomedical and Life Sciences, University of Glasgow, Glasgow G12 8QQ, UK.
| | | | | |
Collapse
|
32
|
|
33
|
Dave KR, DeFazio RA, Raval AP, Torraco A, Saul I, Barrientos A, Perez-Pinzon MA. Ischemic preconditioning targets the respiration of synaptic mitochondria via protein kinase C epsilon. J Neurosci 2008; 28:4172-82. [PMID: 18417696 PMCID: PMC2678917 DOI: 10.1523/jneurosci.5471-07.2008] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/26/2007] [Revised: 02/29/2008] [Accepted: 03/02/2008] [Indexed: 01/01/2023] Open
Abstract
In the brain, ischemic preconditioning (IPC) diminishes mitochondrial dysfunction after ischemia and confers neuroprotection. Activation of epsilon protein kinase C (epsilonPKC) has been proposed to be a key neuroprotective pathway during IPC. We tested the hypothesis that IPC increases the levels of epsilonPKC in synaptosomes from rat hippocampus, resulting in improved synaptic mitochondrial respiration. Preconditioning significantly increased the level of hippocampal synaptosomal epsilonPKC to 152% of sham-operated animals at 2 d of reperfusion, the time of peak neuroprotection. We tested the effect of epsilonPKC activation on hippocampal synaptic mitochondrial respiration 2 d after preconditioning. Treatment with the specific epsilonPKC activating peptide, tat-psiepsilonRACK (tat-psiepsilon-receptor for activated C kinase), increased the rate of oxygen consumption in the presence of substrates for complexes I, II, and IV to 157, 153, and 131% of control (tat peptide alone). In parallel, we found that epsilonPKC activation in synaptosomes from preconditioned animals resulted in altered levels of phosphorylated mitochondrial respiratory chain proteins: increased serine and tyrosine phosphorylation of 18 kDa subunit of complex I, decreased serine phosphorylation of FeS protein in complex III, increased threonine phosphorylation of COX IV (cytochrome oxidase IV), increased mitochondrial membrane potential, and decreased H2O2 production. In brief, ischemic preconditioning promoted significant increases in the level of synaptosomal epsilonPKC. Activation of epsilonPKC increased synaptosomal mitochondrial respiration and phosphorylation of mitochondrial respiratory chain proteins. We propose that, at 48 h of reperfusion after ischemic preconditioning, epsilonPKC is poised at synaptic mitochondria to respond to ischemia either by direct phosphorylation or activation of the epsilonPKC signaling pathway.
Collapse
Affiliation(s)
| | | | | | | | - Isabel Saul
- The Cerebral Vascular Disease Research Center
| | - Antoni Barrientos
- The Cerebral Vascular Disease Research Center
- Department of Neurology and Neuroscience Program, and
- Department of Biochemistry and Molecular Biology and The John T. MacDonald Center for Medical Genetics, Leonard M. Miller School of Medicine, University of Miami, Miami, Florida 33136
| | - Miguel A. Perez-Pinzon
- The Cerebral Vascular Disease Research Center
- Department of Neurology and Neuroscience Program, and
| |
Collapse
|
34
|
Huang L, Cheng HC, Isom R, Chen CS, Levine RA, Pauli BU. Protein kinase Cepsilon mediates polymeric fibronectin assembly on the surface of blood-borne rat breast cancer cells to promote pulmonary metastasis. J Biol Chem 2008; 283:7616-27. [PMID: 18184652 DOI: 10.1074/jbc.m705839200] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Malignant breast cancer cells that have entered the blood circulation from primary mammary fat pad tumors or are grown in end-over-end suspension culture assemble a characteristic, multi-globular polymeric fibronectin (polyFn) coat on their surfaces. Surface polyFn is critical for pulmonary metastasis, presumably by facilitating lung vascular arrest via endothelial dipeptidylpeptidase IV (CD26). Here, we show that cell-surface polyFn assembly is initiated by the state of suspension, is dependent upon the synthesis and secretion of cellular Fn, and is augmented in a dose- and time-dependent manner by plasma Fn. PolyFn assembly is regulated by protein kinase Cepsilon (PKCepsilon), which translocates rapidly and in increasing amounts from the cytosol to the plasma membrane and is phosphorylated. PolyFn assembly is impeded by select inhibitors of this kinase, i.e. bisindolylmaleimide I, Ro-32-0432, Gö6983, and Rottlerin, by the phorbol 12-myristate 13-acetate-mediated and time-dependent loss of PKCepsilon protein and decreased plasma membrane translocation, and more specifically, by stable transfection of lung-metastatic MTF7L breast cancer cells with small interfering RNA-PKCepsilon and dominant-negative PKCepsilon constructs (e.g. RD-PKCepsilon). The inability to assemble a cell surface-associated polyFn coat by knockdown of endogenous Fn or PKCepsilon impedes cancer cells from metastasis to the lungs. The present studies identify a novel regulatory mechanism for polyFn assembly on blood-borne breast cancer cells and depict its effect on pulmonary metastasis.
Collapse
Affiliation(s)
- Lynn Huang
- Cancer Cell Biology Laboratories, Department of Molecular Medicine, Cornell University, Ithaca, New York 14853, USA
| | | | | | | | | | | |
Collapse
|
35
|
Kim E, Raval AP, Defazio RA, Perez-Pinzon MA. Ischemic preconditioning via epsilon protein kinase C activation requires cyclooxygenase-2 activation in vitro. Neuroscience 2007; 145:931-41. [PMID: 17307294 PMCID: PMC2153455 DOI: 10.1016/j.neuroscience.2006.12.063] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2006] [Revised: 12/21/2006] [Accepted: 12/23/2006] [Indexed: 11/23/2022]
Abstract
The signaling pathway of cyclooxygenase-2 (COX-2) induction following ischemic preconditioning (IPC) in brain remains undefined. To determine role of COX-2 in ischemic preconditioning, we used two in vitro models: mixed cortical neuron/astrocyte cell cultures and organotypic hippocampal slice cultures. We simulated IPC by exposing cell or slice cultures to 1 h or 15 min of oxygen/glucose deprivation (OGD), respectively, 48 h prior to ischemia. To mimic ischemia in vitro, we exposed cell or slice cultures to OGD of 4 h or 40 min, respectively. In cell cultures, these experiments revealed that COX-2 induction peaked at 24 h following IPC in cell culture. Inhibition of COX-2 activation with 50 microM NS-398 (a COX-2 selective inhibitor) abolished IPC-mediated neuroprotection in both in vitro models. Next, we tested whether epsilon protein kinase C (epsilonPKC) and extracellular signal regulated kinase 1/2 (ERK1/2) activation was involved in IPC-mediated neuroprotection and COX-2 expression in cell culture. Cell cultures were treated with an epsilonPKC-specific activating peptide (psiepsilonRACK, 100 nM) for 1 h, and 48 h later were exposed to OGD. epsilonPKC activation increased ERK1/2 phosphorylation and COX-2 induction and conferred neuroprotection similar to IPC. Additionally, inhibition of either epsilonPKC or ERK1/2 activation abolished COX-2 expression and neuroprotection due to ischemic preconditioning. These results demonstrate a crucial role for the epsilonPKC-->ERK1/2-->COX-2 pathway in the induction of neuroprotection via ischemic preconditioning.
Collapse
Affiliation(s)
- E Kim
- Cerebral Vascular Disease Research Center, Department of Neurology and Neuroscience Program, University of Miami Miller School of Medicine, Miami, FL 33101, USA
| | | | | | | |
Collapse
|
36
|
Rogel A, Bromberg Y, Sperling O, Zoref-Shani E. The neuroprotective adenosine-activated signal transduction pathway involves activation of phospholipase C. NUCLEOSIDES NUCLEOTIDES & NUCLEIC ACIDS 2007; 25:1283-6. [PMID: 17065107 DOI: 10.1080/15257770600890939] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
We have demonstrated before that exposure of neuronal cultures to poisoning by iodoacetic acid (IAA) followed by "reperfusion" (IAA-R insult), results in severe cytotoxicity, which could be markedly attenuated by prior activation of the adenosine A1 receptors. We also have demonstrated that adenosine activates a signal transduction pathway (STP), which involves activation of PKC epsilon and opening of KATP channels. Here, we provide proof for the involvement also of phospholipase C (PLC) in the neuronal protective adenosine-activated STP. R-PIA, a specific A1 adenosine receptor agonist, was found to enhance neuronal PLC activity and protect against the IAA-R insult. The PLC inhibitor U73122, abrogated both R-PIA-induced effects. These results demonstrate that activation of PLC is a vital step in the neuronal protective adenosine-induced STP.
Collapse
Affiliation(s)
- A Rogel
- Department of Human Molecular Genetics and Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | |
Collapse
|
37
|
Shimohata T, Zhao H, Steinberg GK. Epsilon PKC may contribute to the protective effect of hypothermia in a rat focal cerebral ischemia model. Stroke 2007; 38:375-80. [PMID: 17204679 DOI: 10.1161/01.str.0000254616.78387.ee] [Citation(s) in RCA: 42] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BACKGROUND AND PURPOSE Protein kinase C epsilon (epsilonPKC) has been implicated as a neuroprotectant in vitro. We studied epsilonPKC activation (by its localization and proteolysis) in a rodent stroke model and correlated the effects of hypothermia with epsilonPKC activity after cerebral ischemia. METHODS Rats were subjected to permanent distal middle cerebral artery occlusion plus 1 hour of bilateral common carotid artery occlusion. Body temperatures were maintained at 37 degrees C or 30 degrees C during common carotid artery occlusion. Brains were harvested at 10 minutes, 4 hours, and 24 hours after common carotid artery release, and the cortex corresponding to the ischemic core and penumbra was dissected. epsilonPKC localization after stroke was assessed by Western blot and immunofluorescence microscopy. A caspase-3 inhibitor was used to test whether epsilonPKC cleavage is caspase dependent. RESULTS epsilonPKC in the membrane fraction and whole-protein homogenates decreased moderately in the penumbra but decreased markedly in the ischemic core. Hypothermia blocked this decrease in both the ischemic core and penumbra. Confocal microscopy confirmed that neuronal epsilonPKC expression decreased in the ischemic core at 4 hours after reperfusion, and this loss was prevented by hypothermia. Two carboxyl-terminal cleavage products of epsilonPKC with molecular masses of 43 and 35 kDa were detected. Although the protein band of 43 kDa decreased after stroke, the 35-kDa band increased. Such changes were not dependent on caspase-3. However, hypothermia blocked changes in the cleavage form of 35 kDa but not 43 kDa after stroke. CONCLUSIONS Moderate hypothermia preserves epsilonPKC activity after stroke.
Collapse
Affiliation(s)
- Takayoshi Shimohata
- Department of Neurosurgery, Stanford University School of Medicine, 300 Pasteur Dr R200, Stanford, CA 94305-5327, USA
| | | | | |
Collapse
|
38
|
Jia J, Wang X, Li H, Han S, Zu P, Li J. Activations of nPKCε and ERK1/2 Were Involved in Oxygen-Glucose Deprivation-induced Neuroprotection via NMDA Receptors in Hippocampal Slices of Mice. J Neurosurg Anesthesiol 2007; 19:18-24. [PMID: 17198096 DOI: 10.1097/01.ana.0000211020.88431.e2] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Accumulated reports have suggested that activation of protein kinase C (PKC) isoforms may involve the activation of extracellular signal-regulated kinases (ERKs) in the neuronal response to ischemic/hypoxic stimuli. We have previously demonstrated that the membrane translocation of novel PKC (nPKC) epsilon increased in the early phase of cerebral ischemic/hypoxic preconditioning of mice. In this study, we used Western blot analysis and propidium iodide stain to determine whether the activations of nPKCepsilon and ERKs were involved in oxygen-glucose deprivation (OGD)-induced neuroprotection via N-methyl-D-aspartate (NMDA) receptors. The hippocampal slices of mice were exposed to OGD for 10 (OGD10) or 45 minutes (OGD45) to mimic mild (causing ischemic/hypoxic preconditioning) and severe (causing severe OGD) ischemia/hypoxia, respectively. We found that OGD10-induced nPKCepslilon membrane translocation was mediated by NMDA receptors, and both OGD10 and NMDA (1 microM, 30 min) pretreatment could protect Cornu Ammonis region 1 neurons against the subsequent severe OGD45. In addition, nPKCepsilon translocation inhibitor, epsilonV1-2 (1 microM, 30 min), and ERKs upstream mitogen-activated protein/extracellular signal regulated kinase kinase inhibitor, PD-98059 (20 microM, 30 min), could significantly inhibit OGD10 and NMDA-induced neuroprotection. These results suggest that OGD10-induced neuroprotection against severe OGD45 in the Cornu Ammonis region 1 region of the hippocampal slices was mediated by the activations of NMDA receptors, nPKCepsilon, and the downstream ERKs.
Collapse
Affiliation(s)
- Jun Jia
- Department of Physiology, Institute for Biomedical Science of Pain, Capital Medical University, Beijing 100069, China
| | | | | | | | | | | |
Collapse
|
39
|
Selvatici R, Falzarano S, Franceschetti L, Cavallini S, Marino S, Siniscalchi A. Differential activation of protein kinase C isoforms following chemical ischemia in rat cerebral cortex slices. Neurochem Int 2006; 49:729-36. [PMID: 16963162 DOI: 10.1016/j.neuint.2006.06.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/23/2006] [Accepted: 06/13/2006] [Indexed: 11/23/2022]
Abstract
The aim of the current study was to characterize the effects of chemical ischemia and reperfusion at the transductional level in the brain. Protein kinase C isoforms (alpha, beta(1), beta(2), gamma, delta and epsilon) total levels and their distribution in the particulate and cytosolic compartments were investigated in superfused rat cerebral cortex slices: (i) under control conditions; (ii) immediately after a 5-min treatment with 10mM NaN(3), combined with 2mM 2-deoxyglucose (chemical ischemia); (iii) 1h after chemical ischemia (reperfusion). In control samples, all the PKC isoforms were detected; immediately after chemical ischemia, PKC beta(1), delta and epsilon isoforms total levels (cytosol+particulate) were increased by 2.9, 2.7 and 9.9 times, respectively, while alpha isoform was slightly reduced and gamma isoform was no longer detectable. After reperfusion, the changes displayed by alpha, beta(1), gamma, delta and epsilon were maintained and even potentiated, moreover, an increase in beta(2) (by 41+/-12%) total levels became significant. Chemical ischemia-induced a significant translocation to the particulate compartment of PKC alpha isoform, which following reperfusion was found only in the cytosol. PKC beta(1) and delta isoforms particulate levels were significantly higher both in ischemic and in reperfused samples than in the controls. Conversely, following reperfusion, PKC beta(2) and epsilon isoforms displayed a reduction in their particulate to total level ratios. The intracellular calcium chelator, 1,2-bis(2-aminophenoxy)ethane-N,N,N',N'-tetraacetic acid, 1mM, but not the N-methyl-d-asparate receptor antagonist, MK-801, 1muM, prevented the translocation of beta(1) isoform observed during ischemia. Both drugs were effective in counteracting reperfusion-induced changes in beta(2) and epsilon isoforms, suggesting the involvement of glutamate-induced calcium overload. These findings demonstrate that: (i) PKC isoforms participate differently in neurotoxicity/neuroprotection events; (ii) the changes observed following chemical ischemia are pharmacologically modulable; (iii) the protocol of in vitro chemical ischemia is suitable for drug screening.
Collapse
Affiliation(s)
- Rita Selvatici
- Department of Experimental and Diagnostic Medicine, Medical Genetics Section, University of Ferrara, Via Fossato di Mortara 74, 44100 Ferrara, Italy.
| | | | | | | | | | | |
Collapse
|
40
|
Abstract
The redox environment within neural cells is dependent on a series of redox couples. The glutathione disulfide/ glutathione (GSSG/GSH) redox pair forms the major redox couple in cells and as such plays a critical role in regulating redox-dependent cellular functions. Not only does GSH act as an antioxidant but it also can modulate the activity of a variety of different proteins via S-glutathionylation of cysteine sulfhydryl groups. The thioredoxin system also makes a significant contribution to the redox environment by reducing inter- and intrachain protein disulfide bonds as well as maintaining the activity of important antioxidant enzymes such as peroxiredoxins and methionine sulfoxide reductases. The redox environment affects the activity and function of a number of different protein phosphatases, protein kinases, and transcription factors. The sum of these effects will determine how changes in the redox environment alter overall cellular function, thereby playing a fundamental role in regulating neural cell fate and physiology.
Collapse
Affiliation(s)
- Pamela Maher
- The Salk Institute for Biological Studies, La Jolla, California 92037, USA.
| |
Collapse
|
41
|
Abstract
Background and Purpose—
Stroke is a leading cause of disability and death in the United States, yet limited therapeutic options exist. The need for novel neuroprotective agents has spurred efforts to understand the intracellular signaling pathways that mediate cellular response to stroke. Protein kinase C (PKC) plays a central role in mediating ischemic and reperfusion damage in multiple tissues, including the brain. However, because of conflicting reports, it remains unclear whether PKC is involved in cell survival signaling, or mediates detrimental processes.
Summary of Review—
This review will examine the role of PKC activity in stroke. In particular, we will focus on more recent insights into the PKC isozyme-specific responses in neuronal preconditioning and in ischemia and reperfusion-induced stress.
Conclusion—
Examination of PKC isozyme activities during stroke demonstrates the clinical promise of PKC isozyme-specific modulators for the treatment of cerebral ischemia.
Collapse
Affiliation(s)
- Rachel Bright
- Stanford University School of Medicine, Stanford, CA 94305-5174, USA
| | | |
Collapse
|
42
|
Bell RM, Cave AC, Johar S, Hearse DJ, Shah AM, Shattock MJ. Pivotal role of NOX‐2‐containing NADPH oxidase in early ischemic preconditioning. FASEB J 2005; 19:2037-9. [PMID: 16236999 DOI: 10.1096/fj.04-2774fje] [Citation(s) in RCA: 74] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Reactive oxygen species (ROS)-mediated signaling is implicated in early ischemic preconditioning (PC). A NOX-2-containing NADPH oxidase is a recognized major source of ROS in cardiac myocytes, whose activity is augmented by preconditioning mimetics, such as angiotensin II. We hypothesized that this oxidase is an essential source of ROS in PC. Hearts from wild-type (WT) and NOX-2 knockout (KO) mice were Langendorff perfused and subjected to 35 min ischemia/reperfusion with or without preceding PC or drug treatment. Infarct size was measured by triphenyl tetrazolium chloride staining, and NADPH oxidase activity by lucigenin chemiluminescence. PC significantly attenuated infarct size in WT (26+/-2% vs. control, 38+/-2%, P<0.05) yet was ineffective in KO hearts (33+/-3% vs. control, 34+/-3%). Concomitantly, PC significantly increased NADPH oxidase activity in WT (+41+/-13%; P<0.05), but not in KO (-5+/-18%, P=NS). The ROS scavenger MPG (N-2-mercaptopropionyl glycine, 300 micromol/L) abrogated PC in WT (39+/-2% vs. control, 33+/-1%). CCPA (2-chloro N6 cyclopentyl adenosine, 200 nmol/L), a putative ROS-independent PC trigger, significantly attenuated infarct size in WT, MPG-treated WT and KO hearts (24+/-2, 23+/-1, and 20+/-3%, respectively, P<0.05). Furthermore, CCPA did not augment NADPH oxidase activity over control (+22+/-11%, P=NS). Inhibition of protein kinase C (PKC) with chelerythrine (CHE, 2 micromol/L) completely abrogated both PC (38+/-2% vs. CHE alone, 35+/-2%) and associated increases in oxidase activity (+3+/-10%, P=NS). PKC-dependent activation of a NOX-2-containing NADPH oxidase is pivotally involved in early ischemic PC. However, adenosine receptor activation can trigger a ROS and NOX-2 independent PC pathway.
Collapse
Affiliation(s)
- Robert M Bell
- King's College London, Cardiac Physiology, Cardiovascular Division, The Rayne Institute, St. Thomas' Hospital, London. SE1 7EH, UK.
| | | | | | | | | | | |
Collapse
|
43
|
Kratsovnik E, Bromberg Y, Sperling O, Zoref-Shani E. Oxidative stress activates transcription factor NF-kB-mediated protective signaling in primary rat neuronal cultures. J Mol Neurosci 2005; 26:27-32. [PMID: 15968083 DOI: 10.1385/jmn:26:1:027] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2004] [Accepted: 12/14/2004] [Indexed: 12/31/2022]
Abstract
Activation of transcription factor nuclear factor-kappaB (NF-kappaB) can result in enhanced de novo synthesis of both proteins that confer protection and those that cause death. The present study was undertaken to clarify in primary neuronal cultures the consequences of the oxidative stress-induced activation of NF-kappaB and mediation of death or survival signals. The neuronal cultures were exposed to chemical ischemia (iodoacetic acid), followed by reperfusion (I/R insult). This insult injured the neurons, as manifested in a 7- to 10-fold increase in LDH release, and decreased the cellular content of IkappaBalpha by 55-65 %, indicating NF-kappaB activation. The antioxidants LY231617, melatonin, and sodium salicylate and the antioxidant and inhibitor of NF-kappaB activation pyrrolidine dithiocarbamate, protected the neurons against the insult and prevented the decrease in cellular IkappaBalpha content. In contrast, inhibition of NF-kappaB translocation by SN50 in both uninsulted and insulted neuronal cultures resulted in a 2.9- and 2.4-fold increase in LDH release, respectively. The results indicate that the insult-induced oxidative stress activates transcription factor NF-kappaB associated with induction of protection and suggest that constitutive activation of NF-kappaB under physiological conditions acts to protect the neurons against physiological injury.
Collapse
Affiliation(s)
- Ella Kratsovnik
- Department of Clinical Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | |
Collapse
|
44
|
Libien J, Sacktor TC, Kass IS. Magnesium blocks the loss of protein kinase C, leads to a transient translocation of PKCα and PKCε, and improves recovery after anoxia in rat hippocampal slices. ACTA ACUST UNITED AC 2005; 136:104-11. [PMID: 15893593 DOI: 10.1016/j.molbrainres.2005.01.005] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/26/2004] [Revised: 12/09/2004] [Accepted: 01/09/2005] [Indexed: 12/01/2022]
Abstract
Magnesium is a potent neuroprotective agent against damage to synaptic transmission during cerebral anoxia and reoxygenation. We investigated the mechanisms of anoxic transmission damage and magnesium neuroprotection by examining the response of PKC isoforms to an anoxic insult in the rat hippocampal slice model. A 2-min anoxic period, which resulted in almost complete recovery of synaptic function, did not result in PKC downregulation. In contrast, inducing long-term damage with 10-min anoxia resulted in the downregulation of the conventional PKCs betaI, betaII and gamma immediately after the insult and after 1-h reoxygenation. There was additional loss of PKC(alpha) and PKC(epsilon) after 1-h reoxygenation. Magnesium treatment improved the recovery of synaptic transmission, blocked the loss of PKC and resulted in a transient translocation of PKC(alpha) and PKC(epsilon) to the membrane fraction. Selective downregulation of cPKCs and PKC(epsilon) correlated with permanent damage to synaptic transmission while translocation of PKC(alpha) and PKC(epsilon) correlated with preservation of synaptic function. The mechanisms of magnesium neuroprotection may include altering the PKC response to an anoxic insult.
Collapse
Affiliation(s)
- Jenny Libien
- Department of Physiology and Pharmacology, SUNY Downstate Medical Center, 450 Clarkson Avenue, Brooklyn, NY 11203, USA
| | | | | |
Collapse
|
45
|
Chou WH, Messing RO. Protein Kinase C Isozymes in Stroke. Trends Cardiovasc Med 2005; 15:47-51. [PMID: 15885569 DOI: 10.1016/j.tcm.2005.01.003] [Citation(s) in RCA: 58] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/20/2004] [Revised: 01/27/2005] [Accepted: 01/28/2005] [Indexed: 11/25/2022]
Abstract
Stroke is a devastating neurologic disease and a leading cause of death and disability worldwide. Thrombolytic agents have been used to re-establish circulation in thromboembolic stroke, but their utility is limited by hemorrhage and reperfusion injury. Studies with experimental stroke models, mouse genetics, and selective peptide inhibitors and activators have implicated protein kinase C (PKC) epsilon in ischemic preconditioning and PKCdelta and gamma in tissue injury. PKCdelta, resident both in neutrophils and in the brain, appears particularly essential for reperfusion injury, and recent work using PKCdelta-specific peptide inhibitors suggests that PKCdelta inhibitors could prove useful in attenuating reperfusion injury and improving outcome following thrombolysis.
Collapse
Affiliation(s)
- Wen-Hai Chou
- Ernest Gallo Clinic and Research Center, University of California San Francisco, Emeryville, California 94608, USA
| | | |
Collapse
|
46
|
Siniscalchi A, Marino S, Marani L, Piubello C, Bianchi C, Selvatici R. Early and delayed glutamate effects in rat primary cortical neurons. Neurochem Int 2005; 46:117-25. [PMID: 15627512 DOI: 10.1016/j.neuint.2004.08.001] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2004] [Revised: 08/10/2004] [Accepted: 08/12/2004] [Indexed: 01/29/2023]
Abstract
Glutamate-induced changes in the subcellular distribution of protein kinase C isoforms and in the intracellular calcium concentration were investigated in rat primary cortical neurons. Western blot analysis of protein kinase C isoforms (alpha, beta1, beta2, gamma, delta, epsilon, zeta and theta), performed 30 min after a 10 min treatment with 30 microM glutamate, revealed a decrease in the total beta1 (-24%) and beta2 (-40%) isoform levels, without any significant change in any of the other isozymes. All conventional isoforms translocated to the membrane compartment, while delta, epsilon, zeta and theta; maintained their initial subcellular distribution. Twenty-four hours after glutamate treatment, the total protein kinase C labelling had increased, particularly the epsilon isoform, which accounted for 34% of the total densitometric signal. At this time, protein kinase C beta1, delta, epsilon and zeta isoforms were mainly detected in the membrane compartment, while gamma and theta; signals were displayed almost solely in the cytosol. Basal intracellular calcium concentration (FURA 2 assay) was concentration-dependently increased (maximum effect +77%) 30 min, but not 24h after a 10 min glutamate (10-100 microM) treatment, while the net increase induced by electrical stimulation (10 Hz, 10s) was consistently reduced (maximum effect -64%). The N-methyl-d-aspartate receptor antagonist, MK-801, 1 microM, prevented glutamate action both 30 min and 24 h after treatment, while non-selective protein kinase C inhibitors, ineffective at 30 min, potentiated it at 24 h. These findings show that protein kinase C isoforms are differently activated and involved in the early and delayed glutamate actions, and that the prevailing effect of their activation is neuroprotective.
Collapse
Affiliation(s)
- Anna Siniscalchi
- Department of Clinical and Experimental Medicine, Section of Pharmacology, University of Ferrara, 17, Via Fossato di Mortara, 44100 Ferrara, Italy.
| | | | | | | | | | | |
Collapse
|
47
|
Jung YS, Jung YS, Kim MY, Kim E. Identification of caspase-independent PKCepsilon-JNK/p38 MAPK signaling module in response to metabolic inhibition in H9c2 cells. ACTA ACUST UNITED AC 2004; 54:23-9. [PMID: 15040845 DOI: 10.2170/jjphysiol.54.23] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
To understand the molecular mechanism of ischemia-induced cardiac myocyte cell death, H9c2 cells were studied by chemical hypoxia (CH), using metabolic inhibition buffer. CH suppressed the activities of caspase-3, -8, and -9. c-Jun N-terminal kinase (JNK), and p38 mitogen-activated protein kinase (MAPK) were activated, whereas extracellular regulated kinase (ERK) was inactivated. Only protein kinase Cepsilon (PKCepsilon) among PKC isotypes was translocated to the membrane fraction implying its activation. Moreover, the administration of PKCepsilon inhibitor suppressed the phosphorylations of JNK/p38 MAPK and reduced CH-induced cell death. An administration of JNK/p38 MAPK inhibitors also decreased CH-induced cell deaths, implying JNK/p38 MAPK's causative roles in the deaths. Collectively, this study identified a novel caspase-independent PKCepsilon-JNK/p38 MAPK signaling module induced by CH in cardiac myocytes. Our data show that the PKCepsilon-JNK/p38 MAPK signaling module contributes to CH-induced H9c2 cell death. This contrasts with previous notions, i.e., PKCepsilon's protective effect against ischemic death. Thus our data suggest that PKCepsilon can mediate alternative signals, i.e., beneficiary or deleterious signals, depending on the cell type, intensity, and/or type of injury.
Collapse
Affiliation(s)
- Yong-Sam Jung
- Research Center for Biomedicinal Resources and Division of Life Science, PaiChai University, Daejeon 302-735, Korea
| | | | | | | |
Collapse
|
48
|
Moon CH, Kim MY, Kim MJ, Kim MH, Lee S, Yi KY, Yoo SE, Lee DH, Lim H, Kim HS, Lee SH, Baik EJ, Jung YS. KR-31378, a novel benzopyran analog, attenuates hypoxia-induced cell death via mitochondrial KATP channel and protein kinase C-ɛ in heart-derived H9c2 cells. Eur J Pharmacol 2004; 506:27-35. [PMID: 15588621 DOI: 10.1016/j.ejphar.2004.10.037] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/08/2004] [Revised: 10/13/2004] [Accepted: 10/14/2004] [Indexed: 11/22/2022]
Abstract
A novel compound KR-31378 [(2S,3S,4R)-N''-cyano-N-(6-amino-3,4-dihydro-3-hydroxy-2-methly-2-dimethoxy-methly-2H-benzo-pyran-4-yl)-N-benzylguanidine] has been demonstrated as an anti-ischemic agent in rat heart and brain. Here, we report the effects of this compound on hypoxia-induced cell death and possible signaling pathways in heart-derived H9c2 cells. Treatment with KR-31378 (3-30 microM) 1 h before and during hypoxia significantly reduced hypoxia-induced cell death in a concentration-dependent manner. In addition, increase in hypoxia-induced transferase UTP nick end labeling (TUNEL)-positive cells was reduced by KR-31378, suggesting its antiapoptotic potential in H9c2 cells. The protective effect conferred by KR-31378 (10 microM) was abolished by cotreatment with 5-hydroxydecanoate (5HD), a specific blocker of the mitochondrial KATP (mtKATP) channel, but not by HMR-1883 (1-[[5-[2-(5-chloro-o-anisamido)ethyl]-methoxyphenyl]sulfonyl]-3-methylthiourea), a specific blocker of the sarcolemmal KATP channel. We observed that the treatment with KR-31378 could increase the expression of protein kinase C (PKC)-epsilon protein, but not other PKC isotypes (-alpha, -beta, -delta, -zeta), in the particulate fraction. This increased level of PKC-epsilon was sustained during the hypoxic period up to 8 h. In addition, our results showed that treatment with KR-31378 induced the expression of PKC-epsilon mRNA as early as 15 min after the treatment. A specific inhibitor for PKC-epsilon isoform, epsilonV1-2, completely blocked the protective effect of KR-31378 against hypoxia-induced cell death. In conclusion, our results suggest that KR-31378 can protect cultured H9c2 cells from hypoxia-induced death via the mtKATP channel and PKC-epsilon.
Collapse
Affiliation(s)
- Chang-Hyun Moon
- Department of Physiology, School of Medicine Ajou University, #5 Woncheon-dong, Suwon 442-749, South Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Rogel A, Bromberg Y, Sperling O, Zoref-Shani E. Phospholipase C is involved in the adenosine-activated signal transduction pathway conferring protection against iodoacetic acid-induced injury in primary rat neuronal cultures. Neurosci Lett 2004; 373:218-21. [PMID: 15619546 DOI: 10.1016/j.neulet.2004.10.012] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2004] [Revised: 08/15/2004] [Accepted: 10/04/2004] [Indexed: 11/28/2022]
Abstract
We have demonstrated before that exposure of neuronal cultures to poisoning by iodoacetic acid, followed by "reperfusion" (iodoacetate-"reperfusion" insult; IAA-R insult), results in severe cytotoxicity. This insult was found to be associated with ATP depletion and generation of reactive oxygen species. The cultured neurons could be protected against the insult by activation of the adenosine A1 receptors and by presence of antioxidants. Previous studies in our laboratory demonstrated that the adenosine-activated signal transduction pathway (Ado-STP) conferring protection against the IAA-R insult, involves activation of protein kinase C-epsilon (PKCepsilon) and opening of ATP sensitive potassium (K(ATP)) channels. In this respect, the adenosine-activated protective mechanism against the IAA-R insult is similar to the Ado-STP in the neurons and in cardiomyocytes against ischemia-reperfusion injury. Phospholipase C (PLC) is an additional component demonstrated recently to participate in the myocardial Ado-STP protecting against ischemia-reperfusion. Here we provide proof for the involvement of PLC also in the neuronal Ado-STP protecting against the IAA-R insult. Primary rat neuronal cultures were exposed to the IAA-R insult. The neurons could be protected against this insult by activation of the adenosine A1 receptors by N6-(R)-phenylisopropyladenosine (R-PIA), a specific A1 adenosine receptor agonist. Exposure of the cultures to the PLC inhibitor U73122, abrogated the protection. The exposure of the cultures to R-PIA was found to enhance PLC activity, an effect that could be abrogated by presence of U73122. The R-PIA-induced increase in PLC activity was short-lived, in the range of minutes. These results demonstrate that activation of PLC is a vital step in the neuronal protective Ado-STP, but that it does not contribute directly to the relatively long time window of the protection signal shown previously to characterize the neuronal mechanism. The results also support the suggestion that the Ado-STP protecting against the IAA-R insult and that protecting against ischemia-reperfusion may represent the same mechanism.
Collapse
Affiliation(s)
- Amotz Rogel
- Department of Clinical Biochemistry, Sackler Faculty of Medicine, Tel Aviv University, Tel Aviv, Israel
| | | | | | | |
Collapse
|
50
|
Wang J, Bright R, Mochly-Rosen D, Giffard RG. Cell-specific role for epsilon- and betaI-protein kinase C isozymes in protecting cortical neurons and astrocytes from ischemia-like injury. Neuropharmacology 2004; 47:136-45. [PMID: 15165841 DOI: 10.1016/j.neuropharm.2004.03.009] [Citation(s) in RCA: 37] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2003] [Revised: 02/18/2004] [Accepted: 03/12/2004] [Indexed: 11/17/2022]
Abstract
Activation of epsilon protein kinase C (epsilonPKC) has been shown to protect cardiac myocytes against ischemia and reperfusion injury. However, the role of PKC in ischemic brain injury is less well defined. Western blot analysis of murine neurons and astrocytes in primary culture demonstrated epsilon- and betaIPKC expression in both cell types. Activation of epsilonPKC increased in neuronal cultures in response to the ischemia-like insult of oxygen-glucose deprivation (OGD). Isozyme-specific peptide activators or inhibitors of PKC were applied at various times before, during and after the OGD period. Neuron-astrocyte mixed cultures pretreated with a selective epsilonPKC activator peptide showed a significant reduction in neuronal injury after OGD and reperfusion, compared to cultures pretreated with control peptide. The epsilonPKC activator peptide counteracted the increased damage induced by pretreatment with the epsilonPKC-selective inhibitor peptide in relatively pure neuronal cultures subjected to OGD. Neither epsilonPKC activator nor inhibitor peptides affected injury of neurons when applied after OGD onset. In contrast, the betaIPKC-selective inhibitor peptide increased injury in astrocyte cultures exposed to OGD at all application times tested. Our data demonstrate protection of neurons by selective activation of epsilonPKC but enhanced astrocyte cell death with selective inhibition of betaIPKC. Thus PKC isozymes exhibit cell type-specific effects on ischemia-like injury.
Collapse
Affiliation(s)
- Jian Wang
- Department of Anesthesia, School of Medicine, Stanford University, Stanford, CA 94305-5117, USA
| | | | | | | |
Collapse
|