1
|
Song HJ, Kim JE, Jin YJ, Roh YJ, Seol A, Kim TR, Park KH, Park ES, An BS, Yang SY, Seo S, Jo SM, Jung YS, Hwang DY. Complement C3-Deficiency-Induced Constipation in FVB/N-C3 em1Hlee/Korl Knockout Mice Was Significantly Relieved by Uridine and Liriope platyphylla L. Extracts. Int J Mol Sci 2023; 24:15757. [PMID: 37958740 PMCID: PMC10649790 DOI: 10.3390/ijms242115757] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2023] [Revised: 10/22/2023] [Accepted: 10/27/2023] [Indexed: 11/15/2023] Open
Abstract
Complement component 3 (C3) deficiency has recently been known as a cause of constipation, without studies on the therapeutic efficacy. To evaluate the therapeutic agents against C3-deficiency-induced constipation, improvements in the constipation-related parameters and the associated molecular mechanisms were examined in FVB/N-C3em1Hlee/Korl knockout (C3 KO) mice treated with uridine (Urd) and the aqueous extract of Liriope platyphylla L. (AEtLP) with laxative activity. The stool parameters and gastrointestinal (GI) transit were increased in Urd- and AEtLP-treated C3 KO mice compared with the vehicle (Veh)-treated C3 KO mice. Urd and AEtLP treatment improved the histological structure, junctional complexes of the intestinal epithelial barrier (IEB), mucin secretion ability, and water retention capacity. Also, an improvement in the composition of neuronal cells, the regulation of excitatory function mediated via the 5-hydroxytryptamine (5-HT) receptors and muscarinic acetylcholine receptors (mAChRs), and the regulation of the inhibitory function mediated via the neuronal nitric oxide synthase (nNOS) and inducible NOS (iNOS) were detected in the enteric nervous system (ENS) of Urd- and AEtLP-treated C3 KO mice. Therefore, the results of the present study suggest that C3-deficiency-induced constipation can improve with treatment with Urd and AEtLP via the regulation of the mucin secretion ability, water retention capacity, and ENS function.
Collapse
Affiliation(s)
- Hee-Jin Song
- Department of Biomaterials Science (BK21 FOUR Program)/Life and Industry Convergence Research Institute/Laboratory Animals Resources Center, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea; (H.-J.S.); (J.-E.K.); (Y.-J.J.); (Y.-J.R.); (A.S.); (T.-R.K.); (K.-H.P.); (E.-S.P.); (B.-S.A.); (S.-Y.Y.); (S.S.); (S.-M.J.)
| | - Ji-Eun Kim
- Department of Biomaterials Science (BK21 FOUR Program)/Life and Industry Convergence Research Institute/Laboratory Animals Resources Center, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea; (H.-J.S.); (J.-E.K.); (Y.-J.J.); (Y.-J.R.); (A.S.); (T.-R.K.); (K.-H.P.); (E.-S.P.); (B.-S.A.); (S.-Y.Y.); (S.S.); (S.-M.J.)
| | - You-Jeong Jin
- Department of Biomaterials Science (BK21 FOUR Program)/Life and Industry Convergence Research Institute/Laboratory Animals Resources Center, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea; (H.-J.S.); (J.-E.K.); (Y.-J.J.); (Y.-J.R.); (A.S.); (T.-R.K.); (K.-H.P.); (E.-S.P.); (B.-S.A.); (S.-Y.Y.); (S.S.); (S.-M.J.)
| | - Yu-Jeong Roh
- Department of Biomaterials Science (BK21 FOUR Program)/Life and Industry Convergence Research Institute/Laboratory Animals Resources Center, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea; (H.-J.S.); (J.-E.K.); (Y.-J.J.); (Y.-J.R.); (A.S.); (T.-R.K.); (K.-H.P.); (E.-S.P.); (B.-S.A.); (S.-Y.Y.); (S.S.); (S.-M.J.)
| | - Ayun Seol
- Department of Biomaterials Science (BK21 FOUR Program)/Life and Industry Convergence Research Institute/Laboratory Animals Resources Center, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea; (H.-J.S.); (J.-E.K.); (Y.-J.J.); (Y.-J.R.); (A.S.); (T.-R.K.); (K.-H.P.); (E.-S.P.); (B.-S.A.); (S.-Y.Y.); (S.S.); (S.-M.J.)
| | - Tae-Ryeol Kim
- Department of Biomaterials Science (BK21 FOUR Program)/Life and Industry Convergence Research Institute/Laboratory Animals Resources Center, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea; (H.-J.S.); (J.-E.K.); (Y.-J.J.); (Y.-J.R.); (A.S.); (T.-R.K.); (K.-H.P.); (E.-S.P.); (B.-S.A.); (S.-Y.Y.); (S.S.); (S.-M.J.)
| | - Ki-Ho Park
- Department of Biomaterials Science (BK21 FOUR Program)/Life and Industry Convergence Research Institute/Laboratory Animals Resources Center, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea; (H.-J.S.); (J.-E.K.); (Y.-J.J.); (Y.-J.R.); (A.S.); (T.-R.K.); (K.-H.P.); (E.-S.P.); (B.-S.A.); (S.-Y.Y.); (S.S.); (S.-M.J.)
| | - Eun-Seo Park
- Department of Biomaterials Science (BK21 FOUR Program)/Life and Industry Convergence Research Institute/Laboratory Animals Resources Center, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea; (H.-J.S.); (J.-E.K.); (Y.-J.J.); (Y.-J.R.); (A.S.); (T.-R.K.); (K.-H.P.); (E.-S.P.); (B.-S.A.); (S.-Y.Y.); (S.S.); (S.-M.J.)
| | - Beum-Soo An
- Department of Biomaterials Science (BK21 FOUR Program)/Life and Industry Convergence Research Institute/Laboratory Animals Resources Center, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea; (H.-J.S.); (J.-E.K.); (Y.-J.J.); (Y.-J.R.); (A.S.); (T.-R.K.); (K.-H.P.); (E.-S.P.); (B.-S.A.); (S.-Y.Y.); (S.S.); (S.-M.J.)
| | - Seung-Yun Yang
- Department of Biomaterials Science (BK21 FOUR Program)/Life and Industry Convergence Research Institute/Laboratory Animals Resources Center, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea; (H.-J.S.); (J.-E.K.); (Y.-J.J.); (Y.-J.R.); (A.S.); (T.-R.K.); (K.-H.P.); (E.-S.P.); (B.-S.A.); (S.-Y.Y.); (S.S.); (S.-M.J.)
| | - Sungbaek Seo
- Department of Biomaterials Science (BK21 FOUR Program)/Life and Industry Convergence Research Institute/Laboratory Animals Resources Center, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea; (H.-J.S.); (J.-E.K.); (Y.-J.J.); (Y.-J.R.); (A.S.); (T.-R.K.); (K.-H.P.); (E.-S.P.); (B.-S.A.); (S.-Y.Y.); (S.S.); (S.-M.J.)
| | - Seong-Min Jo
- Department of Biomaterials Science (BK21 FOUR Program)/Life and Industry Convergence Research Institute/Laboratory Animals Resources Center, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea; (H.-J.S.); (J.-E.K.); (Y.-J.J.); (Y.-J.R.); (A.S.); (T.-R.K.); (K.-H.P.); (E.-S.P.); (B.-S.A.); (S.-Y.Y.); (S.S.); (S.-M.J.)
| | - Young-Suk Jung
- College of Pharmacy, Pusan National University, Busan 46241, Republic of Korea;
| | - Dae-Youn Hwang
- Department of Biomaterials Science (BK21 FOUR Program)/Life and Industry Convergence Research Institute/Laboratory Animals Resources Center, College of Natural Resources and Life Science, Pusan National University, Miryang 50463, Republic of Korea; (H.-J.S.); (J.-E.K.); (Y.-J.J.); (Y.-J.R.); (A.S.); (T.-R.K.); (K.-H.P.); (E.-S.P.); (B.-S.A.); (S.-Y.Y.); (S.S.); (S.-M.J.)
| |
Collapse
|
2
|
Nicoll RA, Schulman H. Synaptic memory and CaMKII. Physiol Rev 2023; 103:2877-2925. [PMID: 37290118 PMCID: PMC10642921 DOI: 10.1152/physrev.00034.2022] [Citation(s) in RCA: 47] [Impact Index Per Article: 23.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2022] [Revised: 04/26/2023] [Accepted: 04/30/2023] [Indexed: 06/10/2023] Open
Abstract
Ca2+/calmodulin-dependent protein kinase II (CaMKII) and long-term potentiation (LTP) were discovered within a decade of each other and have been inextricably intertwined ever since. However, like many marriages, it has had its up and downs. Based on the unique biochemical properties of CaMKII, it was proposed as a memory molecule before any physiological linkage was made to LTP. However, as reviewed here, the convincing linkage of CaMKII to synaptic physiology and behavior took many decades. New technologies were critical in this journey, including in vitro brain slices, mouse genetics, single-cell molecular genetics, pharmacological reagents, protein structure, and two-photon microscopy, as were new investigators attracted by the exciting challenge. This review tracks this journey and assesses the state of this marriage 40 years on. The collective literature impels us to propose a relatively simple model for synaptic memory involving the following steps that drive the process: 1) Ca2+ entry through N-methyl-d-aspartate (NMDA) receptors activates CaMKII. 2) CaMKII undergoes autophosphorylation resulting in constitutive, Ca2+-independent activity and exposure of a binding site for the NMDA receptor subunit GluN2B. 3) Active CaMKII translocates to the postsynaptic density (PSD) and binds to the cytoplasmic C-tail of GluN2B. 4) The CaMKII-GluN2B complex initiates a structural rearrangement of the PSD that may involve liquid-liquid phase separation. 5) This rearrangement involves the PSD-95 scaffolding protein, α-amino-3-hydroxy-5-methyl-4-isoxazolepropionic acid receptors (AMPARs), and their transmembrane AMPAR-regulatory protein (TARP) auxiliary subunits, resulting in an accumulation of AMPARs in the PSD that underlies synaptic potentiation. 6) The stability of the modified PSD is maintained by the stability of the CaMKII-GluN2B complex. 7) By a process of subunit exchange or interholoenzyme phosphorylation CaMKII maintains synaptic potentiation in the face of CaMKII protein turnover. There are many other important proteins that participate in enlargement of the synaptic spine or modulation of the steps that drive and maintain the potentiation. In this review we critically discuss the data underlying each of the steps. As will become clear, some of these steps are more firmly grounded than others, and we provide suggestions as to how the evidence supporting these steps can be strengthened or, based on the new data, be replaced. Although the journey has been a long one, the prospect of having a detailed cellular and molecular understanding of learning and memory is at hand.
Collapse
Affiliation(s)
- Roger A Nicoll
- Department of Cellular and Molecular Pharmacology, University of California at San Francisco, San Francisco, California, United States
| | - Howard Schulman
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California, United States
- Panorama Research Institute, Sunnyvale, California, United States
| |
Collapse
|
3
|
Lee DS, Kim TH, Park H, Kim JE. PDI augments kainic acid-induced seizure activity and neuronal death by inhibiting PP2A-GluA2-PICK1-mediated AMPA receptor internalization in the mouse hippocampus. Sci Rep 2023; 13:13927. [PMID: 37626185 PMCID: PMC10457386 DOI: 10.1038/s41598-023-41014-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2023] [Accepted: 08/20/2023] [Indexed: 08/27/2023] Open
Abstract
Protein disulfide isomerase (PDI) is a redox-active enzyme and also serves as a nitric oxide donor causing S-nitrosylation of cysteine residues in various proteins. Although PDI knockdown reduces α-amino-3-hydroxy-5-methylisoxazole-4-propionic acid receptor (AMPAR)-mediated neuronal activity, the underlying mechanisms are largely unknown. In the present study, we found that under physiological condition PDI knockdown increased CaMKII activity (phosphorylation) in the mouse hippocampus. However, PDI siRNA inhibited protein phosphatase (PP) 2A-mediated GluA2 S880 dephosphorylation by increasing PP2A oxidation, independent of S-nitrosylation. PDI siRNA also enhanced glutamate ionotropic receptor AMPA type subunit 1 (GluA1) S831 and GluA2 S880, but not GluA1 S845 and GluA2 Y869/Y873/Y876 phosphorylations, concomitant with the enhanced protein interacting with C kinase 1 (PICK1)-mediated AMPAR internalization. Furthermore, PDI knockdown attenuated seizure activity and neuronal damage in response to kainic acid (a non-desensitizing agonist of AMPAR). Therefore, these findings suggest that PDI may regulate surface AMPAR expression through PP2A-GluA2-PICK1 signaling pathway, and that PDI may be one of the therapeutic targets for epilepsy via AMPAR internalization without altering basal neurotransmission.
Collapse
Affiliation(s)
- Duk-Shin Lee
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, Kangwon-do, 24252, South Korea
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Tae-Hyun Kim
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, Kangwon-do, 24252, South Korea
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Hana Park
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, Kangwon-do, 24252, South Korea
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea
| | - Ji-Eun Kim
- Department of Anatomy and Neurobiology, College of Medicine, Hallym University, Chuncheon, Kangwon-do, 24252, South Korea.
- Institute of Epilepsy Research, College of Medicine, Hallym University, Chuncheon, 24252, South Korea.
| |
Collapse
|
4
|
Maurice T. Protection by sigma-1 receptor agonists is synergic with donepezil, but not with memantine, in a mouse model of amyloid-induced memory impairments. Behav Brain Res 2016; 296:270-278. [PMID: 26386305 DOI: 10.1016/j.bbr.2015.09.020] [Citation(s) in RCA: 51] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2015] [Revised: 09/11/2015] [Accepted: 09/15/2015] [Indexed: 12/14/2022]
Abstract
Drugs activating the sigma-1 (σ1) chaperone protein are anti-amnesic and neuroprotective in neurodegenerative pathologies like Alzheimer's disease (AD). Since these so-called σ1 receptor (σ1R) agonists modulate cholinergic and glutamatergic systems in a variety of physiological responses, we addressed their putative additive/synergistic action in combination with cholinergic or glutamatergic drugs. The selective σ1 agonist PRE-084, or the non-selective σ1 drug ANAVEX2-73 was combined with the acetylcholinesterase inhibitor donepezil or the NMDA receptor antagonist memantine in the nontransgenic mouse model of AD-like memory impairments induced by intracerebroventricular injection of oligomeric Aβ25-35 peptide. Two behavioral tests, spontaneous alternation and passive avoidance response, were used in parallel and both protective and symptomatic effects were examined. After determination of the minimally active doses for each compound, the combinations were tested and the combination index (CI) calculated. Combinations between the σ1 agonists and donepezil showed a synergic protective effect, with CI<1, whereas the combinations with memantine showed an antagonist effect, with CI>1. Symptomatic effects appeared only additive for all combinations, with CI=1. A pharmacological analysis of the PRE-084+donepezil combination revealed that the synergy could be due to an inter-related mechanism involving α7 nicotinic ACh receptors and σ1R. These results demonstrated that σ1 drugs do not only offer a protective potential alone but also in combination with other therapeutic agents. The nature of neuromodulatory molecular chaperone of the σ1R could eventually lead to synergistic combinations.
Collapse
Affiliation(s)
- Tangui Maurice
- Molecular Mechanisms in Neurodegenerative Diseases, MMDN Laboratory, Institut National de la Recherche et de la Santé Médicale, unit 1198, 34095 Montpellier, France; University of Montpellier, 34095 Montpellier, France; Ecole Pratique des Hautes Etudes, 75014 Paris, France.
| |
Collapse
|
5
|
Stratton M, Lee IH, Bhattacharyya M, Christensen SM, Chao LH, Schulman H, Groves JT, Kuriyan J. Activation-triggered subunit exchange between CaMKII holoenzymes facilitates the spread of kinase activity. eLife 2014; 3:e01610. [PMID: 24473075 PMCID: PMC3901001 DOI: 10.7554/elife.01610] [Citation(s) in RCA: 72] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
Abstract
The activation of the dodecameric Ca(2+)/calmodulin dependent kinase II (CaMKII) holoenzyme is critical for memory formation. We now report that CaMKII has a remarkable property, which is that activation of the holoenzyme triggers the exchange of subunits between holoenzymes, including unactivated ones, enabling the calcium-independent phosphorylation of new subunits. We show, using a single-molecule TIRF microscopy technique, that the exchange process is triggered by the activation of CaMKII, and that exchange is modulated by phosphorylation of two residues in the calmodulin-binding segment, Thr 305 and Thr 306. Based on these results, and on the analysis of molecular dynamics simulations, we suggest that the phosphorylated regulatory segment of CaMKII interacts with the central hub of the holoenzyme and weakens its integrity, thereby promoting exchange. Our results have implications for an earlier idea that subunit exchange in CaMKII may have relevance for information storage resulting from brief coincident stimuli during neuronal signaling. DOI: http://dx.doi.org/10.7554/eLife.01610.001.
Collapse
Affiliation(s)
- Margaret Stratton
- Department of Molecular and Cell Biology, Berkeley, Berkeley, United States
| | | | | | | | | | | | | | | |
Collapse
|
6
|
Hing B, Davidson S, Lear M, Breen G, Quinn J, McGuffin P, MacKenzie A. A polymorphism associated with depressive disorders differentially regulates brain derived neurotrophic factor promoter IV activity. Biol Psychiatry 2012; 71:618-26. [PMID: 22265241 PMCID: PMC3712170 DOI: 10.1016/j.biopsych.2011.11.030] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/09/2011] [Revised: 11/23/2011] [Accepted: 11/23/2011] [Indexed: 01/12/2023]
Abstract
BACKGROUND Changes in brain derived neurotrophic factor (BDNF) expression have been associated with mood disorders and cognitive dysfunction. Transgenic models that overexpress or underexpress BDNF demonstrate similar deficits in cognition and mood. We explored the hypothesis that BDNF expression is controlled by balancing the activity of BDNF promoter IV (BP4) with a negative regulatory region containing a polymorphism associated with cognitive dysfunction and mood disorders. METHODS We used comparative genomics, transgenic mouse production, and magnetofection of primary neurons with luciferase reporters and signal transduction agonist treatments to identify novel polymorphic cis-regulatory regions that control BP4 activity. RESULTS We show that BP4 is active in the hippocampus, the cortex, and the amygdala and responds strongly to stimuli such as potassium chloride, lithium chloride, and protein kinase C agonists. We also identified a highly conserved sequence 21 kilobase 5' of BP4 that we called BE5.2, which contains rs12273363, a polymorphism associated with decreased BDNF expression, mood disorders, and cognitive decline. BE5.2 modulated the ability of BP4 to respond to different stimuli. Intriguingly, the rarer disease associated allele, BE5.2(C), acted as a significantly stronger repressor of BP4 activity than the more common BE5.2(T) allele. CONCLUSIONS This study shows that the C allele of rs12273363, which is associated with mood disorder, modulates BP4 activity in an allele-specific manner following cell depolarization or the combined activity of protein kinase A and protein kinase C pathways. The relevance of these findings to the role of BDNF misexpression in mood disorders and cognitive decline is discussed.
Collapse
Affiliation(s)
- Benjamin Hing
- School of Medical Sciences, Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | - Scott Davidson
- School of Medical Sciences, Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | - Marrisa Lear
- School of Medical Sciences, Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| | - Gerome Breen
- Institute of Psychiatry, Kings College London, London, United Kingdom
| | - John Quinn
- Institute of Translational Medicine, University of Liverpool, Liverpool, United Kingdom
| | - Peter McGuffin
- Institute of Psychiatry, Kings College London, London, United Kingdom
| | - Alasdair MacKenzie
- School of Medical Sciences, Institute of Medical Sciences, Foresterhill, University of Aberdeen, Aberdeen, Scotland, United Kingdom
| |
Collapse
|
7
|
Yerlikaya A, Dokudur H. Investigation of the eIF2α phosphorylation mechanism in response to proteasome inhibition in melanoma and breast cancer cells. Mol Biol 2010. [DOI: 10.1134/s0026893310050122] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
|
8
|
Is the lifetime of light-stimulated cGMP phosphodiesterase regulated by recoverin through its regulation of rhodopsin phosphorylation? Behav Brain Sci 2010. [DOI: 10.1017/s0140525x00039522] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
|
9
|
|
10
|
|
11
|
|
12
|
|
13
|
|
14
|
|
15
|
Balasubramanian B, Portillo W, Reyna A, Chen JZ, Moore AN, Dash PK, Mani SK. Nonclassical mechanisms of progesterone action in the brain: I. Protein kinase C activation in the hypothalamus of female rats. Endocrinology 2008; 149:5509-17. [PMID: 18617608 PMCID: PMC2584599 DOI: 10.1210/en.2008-0712] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The modulation of gene regulation by progesterone (P) and its classical intracellular regulation by progestin receptors in the brain, resulting in alterations in physiology and behavior has been well studied. The mechanisms mediating the short latency effects of P are less well understood. Recent studies have revealed rapid nonclassical signaling action of P involving the activation of intracellular signaling pathways. We explored the involvement of protein kinase C (PKC) in P-induced rapid signaling in the ventromedial nucleus of the hypothalamus (VMN) and preoptic area (POA) of the rat brain. Both the Ca2+-independent (basal) PKC activity representing the activation of PKC by the in vivo treatments and the Ca+2-dependent (total) PKC activity assayed in the presence of exogenous cofactors in vitro were determined. A comparison of the two activities demonstrated the strength and temporal status of PKC regulation by steroid hormones in vivo. P treatment resulted in a rapid increase in basal PKC activity in the VMN but not the POA. Estradiol benzoate priming augmented P-initiated increase in PKC basal activity in both the VMN and POA. These increases were inhibited by intracerebroventricular administration of a PKC inhibitor administered 30 min prior to P. The total PKC activity remained unchanged demonstrating maximal PKC activation within 30 min in the VMN. In contrast, P regulation in the POA significantly attenuated total PKC activity +/- estradiol benzoate priming. These rapid changes in P-initiated PKC activity were not due to changes in PKC protein levels or phosphorylation status.
Collapse
Affiliation(s)
- Bhuvana Balasubramanian
- Department of Molecular and Cellular Biology, Baylor College of Medicine, Houston, Texas, USA
| | | | | | | | | | | | | |
Collapse
|
16
|
Guest CB, Deszo EL, Hartman ME, York JM, Kelley KW, Freund GG. Ca2+/calmodulin-dependent kinase kinase alpha is expressed by monocytic cells and regulates the activation profile. PLoS One 2008; 3:e1606. [PMID: 18270593 PMCID: PMC2229650 DOI: 10.1371/journal.pone.0001606] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/23/2007] [Accepted: 01/21/2008] [Indexed: 01/22/2023] Open
Abstract
Macrophages are capable of assuming numerous phenotypes in order to adapt to endogenous and exogenous challenges but many of the factors that regulate this process are still unknown. We report that Ca2+/calmodulin-dependent kinase kinase α (CaMKKα) is expressed in human monocytic cells and demonstrate that its inhibition blocks type-II monocytic cell activation and promotes classical activation. Affinity chromatography with paramagnetic beads isolated an approximately 50 kDa protein from nuclear lysates of U937 human monocytic cells activated with phorbol-12-myristate-13-acetate (PMA). This protein was identified as CaMKKα by mass spectrometry and Western analysis. The function of CaMKKα in monocyte activation was examined using the CaMKKα inhibitors (STO-609 and forskolin) and siRNA knockdown. Inhibition of CaMKKα, enhanced PMA-dependent CD86 expression and reduced CD11b expression. In addition, inhibition was associated with decreased translocation of CaMKKα to the nucleus. Finally, to further examine monocyte activation profiles, TNFα and IL-10 secretion were studied. CaMKKα inhibition attenuated PMA-dependent IL-10 production and enhanced TNFα production indicating a shift from type-II to classical monocyte activation. Taken together, these findings indicate an important new role for CaMKKα in the differentiation of monocytic cells.
Collapse
Affiliation(s)
- Christopher B. Guest
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Eric L. Deszo
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Matthew E. Hartman
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Jason M. York
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Keith W. Kelley
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
| | - Gregory G. Freund
- Division of Nutritional Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Animal Sciences, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of Pathology, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- Department of College of Medicine, University of Illinois at Urbana-Champaign, Urbana, Illinois, United States of America
- *E-mail:
| |
Collapse
|
17
|
Cao G, Gardner A, Westfall TC. Mechanism of dopamine mediated inhibition of neuropeptide Y release from pheochromocytoma cells (PC12 cells). Biochem Pharmacol 2007; 73:1446-54. [PMID: 17286966 PMCID: PMC2709075 DOI: 10.1016/j.bcp.2007.01.003] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2006] [Revised: 12/21/2006] [Accepted: 01/03/2007] [Indexed: 12/30/2022]
Abstract
In rat pheochromocytoma (PC12) cells the dopamine D(2) receptor agonists apomorphine (APO) and n-propylnorapomorphine (NPA) produced a concentration dependent inhibition of K(+)-evoked neuropeptide Y release (NPY-ir). The effect of APO was blocked by the dopamine D(2)-receptor antagonist, eticlopride, but not the D(1)/D(3) or the D(4)/D(2) antagonists, SCH23390 or clozapine, respectively. The D(1)/D(5) receptor agonist, SKF38393 or the D(3) agonists PD128907 and 7-OH DPAT had no effect. Selective N and L-type voltage gated Ca(2+) channel blockers, omega-conotoxin GVIa (Ctx-GVIa) and nifedipine, respectively, produced a concentration dependent inhibition of NPY-ir release but were not additive with APO. The Ca(2+)/calmodulin-dependent protein kinase (CaM kinase) II inhibitor KN-62 produced a concentration-dependent inhibition of NPY-ir release but the combination of KN-62 and APO produced no further inhibition. PMA-mediated protein kinase C stimulation significantly increased both basal and K(+)-evoked release of NPY-ir, and in the presence of PMA APO had no inhibitory effect. The PKC antagonist, chelerythrine, inhibited K(+)-evoked NPY-ir release but was not additive with APO. Neither forskolin-mediated adenylate cyclase activation and the active cAMP analog Sp-cAMPS, nor the adenylate cyclase inhibitor SQ 22536, and the competitive inhibitor of cAMP-dependent protein kinases Rp-cAMPS, had any significant effect on K(+)-evoked NPY-ir release. This suggests the inhibitory effect of APO on K(+)-evoked release of NPY-ir from PC12 cells is most likely mediated through activation of dopamine D(2) receptors leading to direct inhibition of N and L-type voltage gated Ca(2+) channels, or indirect inhibition of PKC, both of which would reduce [Ca(2+)](i) and inactivate CaM kinase.
Collapse
Affiliation(s)
| | - Alice Gardner
- Corresponding author at: Department of Pharmaceutical Sciences, Massachusetts College of Pharmacy and Health Sciences, School of Pharmacy - Worcester, 19 Foster St., Worcester, MA 01608, United States, Tel: + 1 508 373 5665; FAX: + 1 508 890 5618, Email address:
| | | |
Collapse
|
18
|
Juang YT, Wang Y, Solomou EE, Li Y, Mawrin C, Tenbrock K, Kyttaris VC, Tsokos GC. Systemic lupus erythematosus serum IgG increases CREM binding to the IL-2 promoter and suppresses IL-2 production through CaMKIV. J Clin Invest 2005; 115:996-1005. [PMID: 15841182 PMCID: PMC1070410 DOI: 10.1172/jci22854] [Citation(s) in RCA: 180] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/28/2004] [Accepted: 01/05/2005] [Indexed: 01/16/2023] Open
Abstract
Systemic lupus erythematosus (SLE) T cells express high levels of cAMP response element modulator (CREM) that binds to the IL-2 promoter and represses the transcription of the IL-2 gene. This study was designed to identify pathways that lead to increased binding of CREM to the IL-2 promoter in SLE T cells. Ca(2+)/calmodulin-dependent kinase IV (CaMKIV) was found to be increased in the nucleus of SLE T cells and to be involved in the overexpression of CREM and its binding to the IL-2 promoter. Treatment of normal T cells with SLE serum resulted in increased expression of CREM protein, increased binding of CREM to the IL-2 promoter, and decreased IL-2 promoter activity and IL-2 production. This process was abolished when a dominant inactive form of CaMKIV was expressed in normal T cells. The effect of SLE serum resided within the IgG fraction and was specifically attributed to anti-TCR/CD3 autoantibodies. This study identifies CaMKIV as being responsible for the increased expression of CREM and the decreased production of IL-2 in SLE T cells and demonstrates that anti-TCR/CD3 antibodies present in SLE sera can account for the increased expression of CREM and the suppression of IL-2 production.
Collapse
Affiliation(s)
- Yuang-Taung Juang
- Department of Cellular Injury, Walter Reed Army Institute of Research, Silver Spring, Maryland 20910, USA
| | | | | | | | | | | | | | | |
Collapse
|
19
|
Tombes RM, Faison MO, Turbeville JM. Organization and evolution of multifunctional Ca2+/CaM-dependent protein kinase genes. Gene 2003; 322:17-31. [PMID: 14644494 DOI: 10.1016/j.gene.2003.08.023] [Citation(s) in RCA: 163] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The "multi-functional" Ca(2+) and calmodulin-dependent protein kinase, type II (CaMK-II) is an evolutionarily conserved protein. It has been found as a single gene in the horseshoe crab, marine sponge, sea urchin, nematode, and fruit fly, whereas most vertebrates possess four genes (alpha, beta, gamma, and delta). Species from fruit flies to humans encode alternative splice variants which are differentially targeted to phosphorylate diverse downstream targets of Ca(2+) signaling. By comparing known CaMK-II protein and nucleotide sequences, we have now provided evidence for the evolutionary relatedness of CaMK-IIs. Parsimony analyses unambiguously indicate that the four vertebrate CaMK-II genes arose via repeated duplications. Nucleotide phylogenies show consistent but moderate support for the placement of the vertebrate delta CaMK-II as the earliest diverging vertebrate gene. delta CaMK-II is the only gene with both central and C-terminal variable domains and has three to four times more intronic sequence than the other three genes. beta and gamma CaMK-II genes show strong sequence similarity and have comparable exon and intron organization and utilization. alpha CaMK-II is absent from amphibians (Xenopus laevis) and has the most restricted tissue specificity in mammals, whereas beta, gamma, and delta CaMK-IIs are expressed in most tissues. All 38 known mammalian CaMK-II splice variants were compiled with their tissue specificity and exon usage. Some of these variants use alternative 5' and 3' donors within a single exon as well as alternative promoters. These findings serve as an important benchmark for future phylogenetic, developmental, or biochemical studies on this important, conserved, and highly regulated gene family.
Collapse
Affiliation(s)
- Robert M Tombes
- Department of Biology, Virginia Commonwealth University, Richmond, VA 23284-2012, USA.
| | | | | |
Collapse
|
20
|
Rose AJ, Hargreaves M. Exercise increases Ca2+-calmodulin-dependent protein kinase II activity in human skeletal muscle. J Physiol 2003; 553:303-9. [PMID: 14565989 PMCID: PMC2343484 DOI: 10.1113/jphysiol.2003.054171] [Citation(s) in RCA: 135] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
There is evidence in rodents that Ca2+-calmodulin-dependent protein kinase II (CaMKII) activity is higher in contracting skeletal muscle, and this kinase may regulate skeletal muscle function and metabolism during exercise. To investigate the effect of exercise on CaMKII in human skeletal muscle, healthy men (n = 8) performed cycle ergometer exercise for 40 min at 76 +/- 1% peak pulmonary O2 uptake (VO2peak), with skeletal muscle samples taken at rest and after 5 and 40 min of exercise. CaMKII expression and activities were examined by immunoblotting and in vitro kinase assays, respectively. There were no differences in maximal (+ Ca2+, CaM) CaMKII activity during exercise compared with rest. Autonomous (- Ca2+, CaM) CaMKII activity was 9 +/- 1% of maximal at rest, remained unchanged at 5 min, and increased to 17 +/- 1% (P < 0.01) at 40 min. CaMKII autophosphorylation at Thr287 was 50-70% higher during exercise, with no differences in CaMKII expression. The effect of maximal aerobic exercise on CaMKII was also examined (n = 9), with 0.7- to 1.5-fold increases in autonomous CaMKII activity, but no change in maximal CaMKII activity. CaMKIV was not detected in human skeletal muscle. In summary, exercise increases the activity of CaMKII in skeletal muscle, suggesting that it may have a role in regulating skeletal muscle function and metabolism during exercise in humans.
Collapse
Affiliation(s)
- Adam J Rose
- Centre for Physical Activity and Nutrition, School of Health Sciences, Deakin University, Burwood, Australia 3125
| | | |
Collapse
|
21
|
Choe ES, Wang JQ. Regulation of transcription factor phosphorylation by metabotropic glutamate receptor-associated signaling pathways in rat striatal neurons. Neuroscience 2002; 114:557-65. [PMID: 12220559 DOI: 10.1016/s0306-4522(02)00318-4] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
The group I metabotropic glutamate receptors (mGluRs) are positively coupled to phospholipase C. Through phospholipase C, group I mGluR activation increases intracellular concentrations of diacylglycerol which is known as a strong activator of protein kinase C (PKC). This study investigated the putative role of PKC in the regulation of transcription factor phosphorylation induced by group I mGluR activation in the rat striatum in vivo. We found that the group I agonist 3,5-dihydroxyphenylglycine (DHPG) injected into the dorsal striatum (caudate-putamen) increased phosphorylation of the two transcription factors, cAMP response element-binding protein (CREB) and Elk-1, and extracellular signal-regulated kinase 1/2 (ERK1/2) in the injected striatum. Inhibition of PKC with GF109203X significantly attenuated DHPG-stimulated CREB, Elk-1, and ERK1/2 phosphorylation. Activation of PKC with intracaudate injection of 12-O-tetradecanoylphorbol-13-acetate (TPA) mimicked DHPG actions in facilitating the phosphorylation of CREB, Elk-1, and ERK1/2. Blockade of N-methyl-D-aspartate (NMDA) glutamate receptors with the non-competitive antagonist MK801 or the competitive antagonist AP5 attenuated TPA-induced CREB, Elk-1, and ERK1/2 phosphorylation. Similarly, inhibition of Ca(2+)/calmodulin-dependent protein kinases (CaMK) with KN62 also resulted in a significant attenuation of TPA induction of the three phosphoproteins. The data obtained from this study indicate that selective activation of PKC is needed for the group I agonist-induced CREB, Elk-1, and ERK1/2 phosphorylation in striatal neurons. Activated PKC may, at least in part, facilitate the phosphorylation of transcription factors via an NMDA/CaMK-sensitive pathway.
Collapse
Affiliation(s)
- E S Choe
- Division of Pharmacology, School of Pharmacy, University of Missouri-Kansas City, 2411 Holmes Street, M3-C225, Kansas City, MO 64108, USA
| | | |
Collapse
|
22
|
Lin X, Gelman IH. Calmodulin and cyclin D anchoring sites on the Src-suppressed C kinase substrate, SSeCKS. Biochem Biophys Res Commun 2002; 290:1368-75. [PMID: 11820772 DOI: 10.1006/bbrc.2002.6357] [Citation(s) in RCA: 38] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
SSeCKS and its human orthologue, Gravin, are large scaffolding proteins that are thought to facilitate mitogenic control by anchoring key signal mediators such as protein kinase (PK) C, PKA, the plasma membrane associated isoform of alpha-1,4-galactosyltransferase (GalTase), beta2-adrenergic receptor, and cyclins. SSeCKS is also a major PKC substrate and phosphatidylserine-dependent PKC binding protein whose phosphorylation sites shares homology with a site in the MARCKS protein that encodes phosphorylation-sensitive calmodulin (CaM) binding activity. In the present study, we mapped the in vitro binding sites for CaM and cyclins on SSeCKS. Four CaM binding sites were identified by binding assays that conform to the so-called 1-5-10 motif. Notably, CaM binding was antagonized by prephosphorylation of SSeCKS by PKC. We also identified two major cyclin binding (CY) sites that overlap a major PKC phosphorylation site in SSeCKS (Ser(507/515)), and showed that cyclin D binding is attenuated if SSeCKS is prephosphorylated by PKC. These data suggest that the scaffolding activities of SSeCKS are modulated by mitogenically stimulated kinases such as PKC.
Collapse
Affiliation(s)
- Xueying Lin
- Department of Medicine and Ruttenberg Cancer Center, Mount Sinai School of Medicine, Box 1090, One Gustave L. Levy Place, New York, New York 10029-6574, USA
| | | |
Collapse
|
23
|
Hung AC, Sun SH. The P2X(7) receptor-mediated phospholipase D activation is regulated by both PKC-dependent and PKC-independent pathways in a rat brain-derived Type-2 astrocyte cell line, RBA-2. Cell Signal 2002; 14:83-92. [PMID: 11747993 DOI: 10.1016/s0898-6568(01)00230-3] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
The aim of this study was to characterize the regulatory mechanisms of the P2X(7) receptor (P2X(7)R)-mediated phospholipase D (PLD) activation in a rat brain-derived Type-2 astrocyte cell line, RBA-2. A time course study revealed that activation of P2X(7)R resulted in a choline and not phosphorylcholine formation, suggesting that activation of P2X(7)R is associated with the phosphatidylcholine-PLD (PC-PLD) in these cells. GF 109203X, a selective protein kinase C (PKC) inhibitor, partially inhibited the P2X(7)R-mediated PLD activation, while blocking the phorbol 12-myristate 13-acetate (PMA)-stimulated PLD activity. In addition, PMA synergistically activated the P2X(7)R-mediated PLD activity. Furthermore, genistein, a tyrosine kinase inhibitor, blocked the P2X(7)R-activated PLD, while KN62, a Ca(2+)/calmodulin-dependent protein kinase II (CaMKII) inhibitor, was less effective, whereas the mitogen-activated protein kinase (MAPK) inhibitor PD98059 was ineffective. No additive inhibitory effects were found by simultaneous treatment of GF 109203X and KN62 on P2X(7)R-activated PLD. Taken together, these results demonstrate that both PKC-dependent and PKC-independent signaling pathways are involved in the regulation of P2X(7)R-mediated PLD activation. Additionally, CaMKII may participate in the PKC-dependent pathway, and tyrosine kinase may play a pivotal role on both PKC-dependent and PKC-independent pathways in the P2X(7)R-mediated PLD activation in RBA-2 cells.
Collapse
Affiliation(s)
- Amos C Hung
- Institute of Neuroscience, College of Life Science, National Yang Ming University, No. 155, Section 2, Li-Non Street, Shi-Pai, Taipei 112, Taiwan, ROC
| | | |
Collapse
|
24
|
Gardoni F, Bellone C, Cattabeni F, Di Luca M. Protein kinase C activation modulates alpha-calmodulin kinase II binding to NR2A subunit of N-methyl-D-aspartate receptor complex. J Biol Chem 2001; 276:7609-13. [PMID: 11104776 DOI: 10.1074/jbc.m009922200] [Citation(s) in RCA: 92] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The N-methyl-d-aspartate (NMDA) receptor subunits NR2 possess extended intracellular C-terminal domains by which they can directly interact with a large number of postsynaptic density (PSD) proteins involved in synaptic clustering and signaling. We have previously shown that PSD-associated alpha-calmodulin kinase II (alphaCaMKII) binds with high affinity to the C-terminal domain of the NR2A subunit. Here, we show that residues 1412-1419 of the cytosolic tail of NR2A are critical for alphaCaMKII binding, and we identify, by site directed mutagenesis, PKC-dependent phosphorylation of NR2A(Ser(1416)) as a key mechanism in inhibiting alphaCaMKII-binding and promoting dissociation of alphaCaMKII.NR2A complex. In addition, we show that stimulation of PKC activity in hippocampal slices either with phorbol esters or with the mGluRs specific agonist trans-1-amino-1,3- cyclopentanedicarboxylic acid (t-ACPD) decreases alphaCaMKII binding to NMDA receptor complex. Thus, our data provide clues on understanding the molecular basis of a direct cross-talk between alphaCaMKII and PKC pathways in the postsynaptic compartment.
Collapse
Affiliation(s)
- F Gardoni
- Institute of Pharmacological Sciences, University of Milano, via Balzaretti 9, 20133 Milano, Italy.
| | | | | | | |
Collapse
|
25
|
Robidoux J, Simoneau L, Masse A, Lafond J. Activation of L-type calcium channels induces corticotropin-releasing factor secretion from human placental trophoblasts. J Clin Endocrinol Metab 2000; 85:3356-64. [PMID: 10999834 DOI: 10.1210/jcem.85.9.6774] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The ultimate outcome of pregnancy, parturition, is a well orchestrated process in which placental corticotropin-releasing factor (CRF) seems to play an important role. The objective of the present study was to investigate the involvement of L-type calcium channels and calcium-dependent signaling in the depolarization-induced CRF release from human syncytiotrophoblast. The basal secretion of CRF by trophoblastic cells, isolated from human term placenta, was maximal after their functional differentiation, which was monitored by hCG measurements. On the fourth day of culture, the basal CRF secretion of the cells in serum-free medium was linear between 2 and 8 h. Incubation of the trophoblasts with KCl, a depolarizing stimulus, or with Bay K8644, an L-type calcium channel agonist, for 3 or 8 h led to an increase in CRF secretion, but was without effect on its synthesis. This stimulated CRF release was calcium dependent, as it could be prevented by loading cells with 1,2-bis(0-Aminophenoxy)ethane-N,N,N',N'-tetraacetic acid (acetoxymethyl) ester. Furthermore, the KCl-induced CRF secretion involved L-type calcium channels activation, as 2 micromol/L nitrendipine, an L-type specific blocker, abolished the stimulation. In trophoblasts, where we have previously shown calcium-dependent protein kinase C (cPKCs) activity, incubation with Bay K8644 also stimulated calcium calmodulin kinase II (CaMKII) and extracellular regulated kinase activities. In the present study we observed that CaMKII and cPKCs were linked to the Bay K8644-induced secretion of CRF, as only the autocamtide-2 related inhibitory peptide, a CaMKII inhibitor, and Gö6976, an inhibitor of mu and cPKCs partially prevented (30-78%) the activation of CRF release by Bay K8644. The use of PD 098056, an inhibitor of the ERKs kinases, showed no effect on CRF release. Taken together, these results support a depolarization-induced and calcium-dependent exocytotic-like secretion of CRF from human placental trophoblasts. In addition, CaMKII and cPKCs seem to be potential modulators or mediators of these calcium effects on CRF secretion.
Collapse
Affiliation(s)
- J Robidoux
- Programme des Sciences Biomédicales de la Faculté de Médecine, Université de Montréal, Québec, Canada
| | | | | | | |
Collapse
|
26
|
Walaas SI, Sefland I. Modulation of calcium-evoked [3H]noradrenaline release from permeabilized cerebrocortical synaptosomes by the MARCKS protein, calmodulin and the actin cytoskeleton. Neurochem Int 2000; 36:581-93. [PMID: 10771116 DOI: 10.1016/s0197-0186(99)00159-x] [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] [Indexed: 11/24/2022]
Abstract
In order to examine intracellular modulation of CNS catecholamine release, cerebrocortical synaptosomes were prelabeled with [3H]noradrenaline and permeabilized with streptolysin-O in the absence or presence of Ca(2+). Plasma membrane permeabilization allowed efflux of cytosol and left a compartmentalized pool of [3H]noradrenaline intact, approximately 10% of which was released by addition of 10(-5) M Ca(2+). Addition of activators or inhibitors of protein kinase C, as well as inhibitors of Ca(2+)-calmodulin kinase II or calcineurin, failed to change Ca(2+)-induced noradrenaline release. Evoked release from permeabilized synaptosomes deficient in the vesicle-associated phosphoprotein synapsin I was also unchanged. In contrast, addition of a synthetic 'active domain' peptide from the myristoylated, alanine-rich C-kinase substrate (MARCKS) protein increased, while addition of calmodulin decreased Ca(2+)-induced release from the permeabilized synaptosomes, the latter effect being reversed by a peptide inhibitor of calcineurin. Moreover, addition of the actin-destabilizing agent DNase I, as well as antibodies to MARCKS, appeared to increase spontaneous, Ca(2+)-independent release from noradrenergic vesicles. These results indicate that the MARCKS protein may modulate release from permeabilized noradrenergic synaptosomes, possibly by modulating calmodulin levels and/or the actin cytoskeleton.
Collapse
Affiliation(s)
- S I Walaas
- Neurochemical Laboratory, Institute of Basic Medical Sciences, University of Oslo, P.O. Box 1115-Blindern, N-0317, Oslo, Norway.
| | | |
Collapse
|
27
|
Yang CM, Tsai YJ, Pan SL, Lin CC, Wu WB, Wang CC, Huang SC, Chiu CT. Inhibition of bradykinin-induced phosphoinositide hydrolysis and Ca2+ mobilisation by phorbol ester in rat cultured vascular smooth muscle cells. Cell Signal 1999; 11:899-907. [PMID: 10659998 DOI: 10.1016/s0898-6568(99)00061-3] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Regulation of the increase in inositol phosphate (IP) production and intracellular Ca2+ concentration ([Ca2+]i by protein kinase C (PKC) was investigated in cultured rat vascular smooth muscle cells (VSMCs). Pretreatment of VSMCs with phorbol 12-myristate 14-acetate (PMA, 1 microM) for 30 min almost abolished the BK-induced IP formation and Ca2+ mobilisation. This inhibition was reduced after incubating the cells with PMA for 4 h, and within 24 h the BK-induced responses were greater than those of control cells. The concentrations of PMA giving a half-maximal (pEC50) and maximal inhibition of BK induced an increase in [Ca2+]i, were 7.8 +/- 0.3 M and 1 microM, n = 8, respectively. Prior treatment of VSMCs with staurosporine (1 microM), a PKC inhibitor, inhibited the ability of PMA to attenuate BK-induced responses, suggesting that the inhibitory effect of PMA is mediated through the activation of PKC. Paralleling the effect of PMA on the BK-induced IP formation and Ca2+ mobilisation, the translocation and downregulation of PKC isozymes were determined by Western blotting with antibodies against different PKC isozymes. The results revealed that treatment of the cells with PMA for various times, translocation of PKC-alpha, betaI, betaII, delta, epsilon, and zeta isozymes from the cytosol to the membrane were seen after 5 min, 30 min, 2 h, and 4 h of treatment. However, 24-h treatment caused a partial downregulation of these PKC isozymes in both fractions. Treatment of VSMCs with 1 microM PMA for either 1 or 24 h did not significantly change the K(D) and Bmax of the BK receptor for binding (control: K(D) = 1.7 +/- 0.2 nM; Bmax = 47.3 +/- 4.4 fmol/mg protein), indicating that BK receptors are not a site for the inhibitory effect of PMA on BK-induced responses. In conclusion, these results demonstrate that translocation of PKC-alpha, betaI, betaII, delta, epsilon, and zeta induced by PMA caused an attenuation of BK-induced IPs accumulation and Ca2+ mobilisation in VSMCs.
Collapse
MESH Headings
- Animals
- Biological Transport/drug effects
- Bradykinin/antagonists & inhibitors
- Bradykinin/pharmacology
- Calcium Signaling/drug effects
- Cells, Cultured
- Enzyme Activation/drug effects
- Enzyme Inhibitors/pharmacology
- Indoles/pharmacology
- Inositol 1,4,5-Trisphosphate/metabolism
- Isoenzymes/antagonists & inhibitors
- Isoenzymes/metabolism
- Kinetics
- Maleimides/pharmacology
- Muscle, Smooth, Vascular/cytology
- Muscle, Smooth, Vascular/drug effects
- Muscle, Smooth, Vascular/metabolism
- Phosphatidylinositols/metabolism
- Protein Kinase C/antagonists & inhibitors
- Protein Kinase C/metabolism
- Rats
- Rats, Sprague-Dawley
- Staurosporine/pharmacology
- Tetradecanoylphorbol Acetate/pharmacology
Collapse
Affiliation(s)
- C M Yang
- Department of Pharmacology, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan.
| | | | | | | | | | | | | | | |
Collapse
|
28
|
Kobayashi S, Millhorn DE. Stimulation of expression for the adenosine A2A receptor gene by hypoxia in PC12 cells. A potential role in cell protection. J Biol Chem 1999; 274:20358-65. [PMID: 10400659 DOI: 10.1074/jbc.274.29.20358] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The purpose of this study was to examine the regulation of adenosine A2A receptor (A2AR) gene expression during hypoxia in pheochromocytoma (PC12) cells. Northern blot analysis revealed that the A2AR mRNA level was substantially increased after a 3-h exposure to hypoxia (5% O2), which reached a peak at 12 h. Immunoblot analysis showed that the A2AR protein level was also increased during hypoxia. Inhibition of de novo protein synthesis blocked A2AR induction by hypoxia. In addition, removal of extracellular free Ca2+, chelation of intracellular free Ca2+, and pretreatment with protein kinase C inhibitors prevented A2AR induction by hypoxia. Moreover, depletion of protein kinase C activity by prolonged treatment with phorbol 12-myristate 13-acetate significantly inhibited the hypoxic induction of A2AR. A2AR antagonists led to a significant enhancement of A2AR mRNA levels during hypoxia, whereas A2AR agonists caused down-regulation of A2AR expression during hypoxia. This suggests that A2AR regulates its own expression during hypoxia by feedback mechanisms. We further found that activation of A2AR enhances cell viability during hypoxia and also inhibits vascular endothelial growth factor expression in PC12 cells. Thus, increased expression of A2AR during hypoxia might protect cells against hypoxia and may act to inhibit hypoxia-induced angiogenic activity mediated by vascular endothelial growth factor.
Collapse
Affiliation(s)
- S Kobayashi
- Department of Molecular and Cellular Physiology, University of Cincinnati, Cincinnati, Ohio 45267-576, USA
| | | |
Collapse
|
29
|
Famulski KS, Paterson MC. Defective regulation of Ca2+/calmodulin-dependent protein kinase II in gamma-irradiated ataxia telangiectasia fibroblasts. FEBS Lett 1999; 453:183-6. [PMID: 10403399 DOI: 10.1016/s0014-5793(99)00664-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
Recent indirect evidence suggests that a Ca2+/ calmodulin-dependent pathway, which may involve calmodulin-dependent protein kinase II (CaMKII), mediates the S-phase delay manifested by gamma-ray-exposed human fibroblasts. This pathway is severely impaired in ataxia telangiectasia (A-T) cells [Mirzayans et al. (1995) Oncogene 11, 15971. To extend these findings, we assayed CaMKII activity in irradiated normal and A-T fibroblasts. The radiation treatment induced the autonomous activity of the kinase in normal cells. In contrast, this activity was not elevated in either (i) normal cells pretreated with the selective CaMKII antagonist KN-62 or (ii) gamma-irradiated A-T cells. Moreover, A-T fibroblasts, unlike normal cells, failed to mobilize intracellular Ca2+ upon mitogenic stimulation. These findings identify a novel role for CaMKII in radiation-induced signal transduction and suggest its involvement in effecting the S-phase delay. The data also implicate ATM, the product of the gene responsible for A-T, as a key mediator of both intracellular Ca2+ mobilization and CaMKII activation in response not only to genotoxic stress but also to physiological stimuli.
Collapse
Affiliation(s)
- K S Famulski
- Department of Biological and Medical Research, King Faisal Specialist Hospital and Research Centre, Riyadh, Saudi Arabia.
| | | |
Collapse
|
30
|
Haak LL. Metabotropic glutamate receptor modulation of glutamate responses in the suprachiasmatic nucleus. J Neurophysiol 1999; 81:1308-17. [PMID: 10085357 DOI: 10.1152/jn.1999.81.3.1308] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Glutamate is the primary excitatory transmitter in the suprachiasmatic nucleus (SCN). Ionotropic glutamate receptors (iGluRs) mediate transduction of light information from the retina to the SCN, an important circadian clock phase shifting pathway. Metabotropic glutamate receptors (mGluRs) may play a significant modulatory role. mGluR modulation of SCN responses to glutamate was investigated with fura-2 calcium imaging in SCN explant cultures. SCN neurons showed reproducible calcium responses to glutamate, kainate, and N-methyl-D-aspartate (NMDA). Although the type I/II mGluR agonists L-CCG-I and t-ACPD did not evoke calcium responses, they did inhibit kainate- and NMDA-evoked calcium rises. This interaction was insensitive to pertussis toxin. Protein kinase A (PKA) activation by 8-bromo-cAMP significantly reduced iGluR inhibition by mGluR agonists. The inhibitory effect of mGluRs was enhanced by activating protein kinase C (PKC) and significantly reduced in the presence of the PKC inhibitor H7. Previous reports show that L-type calcium channels can be modulated by PKC and PKA. In SCN cells, about one-half of the calcium rise evoked by kainate or NMDA was blocked by the L-type calcium channel antagonist nimodipine. Calcium rises evoked by K+ were used to test whether mGluR inhibition of iGluR calcium rises involved calcium channel modulation. These calcium rises were primarily attributable to activation of voltage-activated calcium channels. PKC activation inhibited K+-evoked calcium rises, but PKC inhibition did not affect L-CCG-I inhibition of these rises. In contrast, 8Br-cAMP had no effect alone but blocked L-CCG-I inhibition. Taken together, these results suggest that activation of mGluRs, likely type II, modulates glutamate-evoked calcium responses in SCN neurons. mGluR inhibition of iGluR calcium rises can be differentially influenced by PKC or PKA activation. Regulation of glutamate-mediated calcium influx could occur at L-type calcium channels, K+ channels, or at GluRs. It is proposed that mGluRs may be important regulators of glutamate responsivity in the circadian system.
Collapse
Affiliation(s)
- L L Haak
- Neurosciences Program, Department of Biological Sciences, Stanford University, Stanford, California 94305-5020, USA
| |
Collapse
|
31
|
Kusama T, Hatama K, Sakurai M, Kizawa Y, Uhl GR, Murakami H. Consensus phosphorylation sites of human GABA(c)/GABArho receptors are not critical for inhibition by protein kinase C activation. Neurosci Lett 1998; 255:17-20. [PMID: 9839716 DOI: 10.1016/s0304-3940(98)00696-x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The mechanism of inhibition of human GABA(C)/GABArho receptors by protein kinase C (PKC) activation was investigated in Xenopus oocytes. Phorbol 12-myristate 13 acetate (PMA), a potent PKC activator, at 25 nM inhibited the currents through GABArho2 receptors, which have one consensus phosphorylation site by PKC in the predicted intracellular loops. The time-courses and amplitudes of inhibition were not significantly different from those occurring through GABArho1 receptors, which have six such sites. The inhibitory effect of PMA was also observed after removing each consensus phosphorylation site in both GABArho1 and rho2 receptors by site-directed mutagenesis. These results suggest that phosphorylation of consensus sites in the intracellular loops is not involved in the inhibition of human GABA(C)/GABArho receptors by PKC activation.
Collapse
Affiliation(s)
- T Kusama
- Department of Physiology and Anatomy, Nihon University College of Pharmacy, Funabashi, Chiba, Japan
| | | | | | | | | | | |
Collapse
|
32
|
Yang CM, Luo SF, Wu WB, Pan SL, Tsai YJ, Chiu CT, Wang CC. Uncoupling of bradykinin-induced phosphoinositide hydrolysis and Ca2+ mobilization by phorbol ester in canine cultured tracheal epithelial cells. Br J Pharmacol 1998; 125:627-36. [PMID: 9831895 PMCID: PMC1571019 DOI: 10.1038/sj.bjp.0702094] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
1. Regulation of the increase in inositol phosphates (IPs) production and intracellular Ca2+ concentration ([Ca2+]i by protein kinase C (PKC) was investigated in canine cultured tracheal epithelial cells (TECs). Stimulation of TECs by bradykinin (BK) led to IPs formation and caused an initial transient [Ca2+]i peak in a concentration-dependent manner. 2. Pretreatment of TECs with phorbol 12-myristate 13-acetate (PMA, 1 microM) for 30 min attenuated the BK-induced IPs formation and Ca2+ mobilization. The maximal inhibition occurred after incubating the cells with PMA for 2 h. 3. The concentrations of PMA that gave half-maximal (pEC50) inhibition of BK-induced IPs accumulation and an increase in [Ca2+]i were 7.07 M and 7.11 M, respectively. Inactive phorbol ester, 4alpha-phorbol 12,13-didecanoate at 1 microM, did not inhibit these responses. Prior treatment of TECs with staurosporine (1 microM), a PKC inhibitor, inhibited the ability of PMA to attenuate BK-induced responses, suggesting that the inhibitory effect of PMA is mediated through the activation of PKC. 4. In parallel with the effect of PMA on the BK-induced IPs formation and Ca2+ mobilization, the translocation and down-regulation of PKC isozymes were determined. Analysis of cell extracts by Western blotting with antibodies against different PKC isozymes revealed that TECs expressed PKC-alpha, betaI, betaII, gamma, delta, epsilon, theta and zeta. With PMA treatment of the cells for various times, translocation of PKC-alpha, betaI, betaII, gamma, delta, epsilon and theta from cytosol to the membrane was seen after 5 min, 30 min, 2 h, and 4 h treatment. However, 6 h treatment caused a partial down-regulation of these PKC isozymes. PKC-zeta was not significantly translocated and down-regulated at any of the times tested. 5. Treatment of TECs with 1 microM PMA for either 30 min or 6 h did not significantly change the KD, and Bmax receptor for BK binding (control: KD=1.7+/-0.3 nM; Bmax=50.5+/-4.9 fmol/mg protein), indicating that BK receptors are not a site for the inhibitory effect of PMA on BK-induced responses. 6. In conclusion, these results suggest that activation of PKC may inhibit the phosphoinositide hydrolysis and consequently attenuate the [Ca2+]i increase or inhibit independently both responses to BK. The translocation of pKC-alpha, betaI, betaII, delta, epsilon, gamma, and theta induced by PMA caused an attenuation of BK-induced IPs accumulation and Ca2+ mobilization in TECs.
Collapse
Affiliation(s)
- C M Yang
- Department of Pharmacology, College of Medicine, Chang Gung University, Kwei-San, Tao-Yuan, Taiwan
| | | | | | | | | | | | | |
Collapse
|
33
|
McGinnis KM, Whitton MM, Gnegy ME, Wang KK. Calcium/calmodulin-dependent protein kinase IV is cleaved by caspase-3 and calpain in SH-SY5Y human neuroblastoma cells undergoing apoptosis. J Biol Chem 1998; 273:19993-20000. [PMID: 9685336 DOI: 10.1074/jbc.273.32.19993] [Citation(s) in RCA: 84] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
We have previously demonstrated cleavage of alpha-spectrin by caspase-3 and calpain during apoptosis in SH-SY5Y neuroblastoma cells (Nath, R., Raser, K. J., Stafford, D., Hajimohammadreza, I., Posner, A., Allen, H., Talanian, R. V., Yuen, P., Gilbertsen, R. B., and Wang, K. K. (1996) Biochem. J. 319, 683-690). We demonstrate here that calcium/calmodulin-dependent protein kinase IV (CaMK IV) is cleaved during apoptosis by caspase-3 and calpain. We challenged SH-SY5Y cells with the pro-apoptotic agent thapsigargin. Western blot analysis revealed major CaMK IV breakdown products of 40, 38, and 33 kDa. Digestion of control SH-SY5Y lysate with purified caspase-3 produced a 38-kDa CaMK IV fragment; digestion with purified calpain produced a major fragment of 40 kDa. Pretreatment with carbobenzoxy-Asp-CH2OC(O)-2,6-dichlorobenzene or Z-Val-Ala-Asp-fluoromethylketone was able to block the caspase-3-mediated production of the 38-kDa fragment both in situ and in vitro. Calpain inhibitor II similarly blocked formation of the calpain-mediated 40-kDa fragment both in situ and in vitro. Digestion of recombinant CaMK IV by other caspase family members revealed that only caspase-3 produces a fragmentation pattern consistent to that seen in situ. The major caspase-3 and calpain cleavage sites are respectively identified as PAPD176*A and CG201*A, both within the CaMK IV catalytic domain. Furthermore, calmodulin-stimulated protein kinase activity decreases within 6 h in thapsigargin-treated SH-SY5Y. The loss of activity precedes cell death.
Collapse
Affiliation(s)
- K M McGinnis
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor, Michigan 48109, USA
| | | | | | | |
Collapse
|
34
|
le Gouvello S, Manceau V, Sobel A. Serine 16 of Stathmin as a Cytosolic Target for Ca2+/Calmodulin-Dependent Kinase II After CD2 Triggering of Human T Lymphocytes. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.3.1113] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
We investigated specific signaling events initiated after T cell triggering through the costimulatory surface receptors CD2 and CD28 as compared with activation via the Ag receptor (TCR/CD3). We therefore followed the phosphorylation of stathmin, a ubiquitous cytoplasmic phosphoprotein proposed as a general relay integrating diverse intracellular signaling pathways through the combinatorial phosphorylation of serines 16, 25, 38, and 63, the likely physiologic substrates for Ca2+/calmodulin (CaM)-dependent kinases, mitogen-activated protein (MAP) kinase, cyclin-dependent kinases (cdks), and protein kinase A, respectively. We addressed the specific protein kinase systems involved in the CD2 pathway of T cell activation through the analysis of stathmin phosphorylation patterns in exponentially growing Jurkat T cells, as revealed by phosphopeptide mapping. Stimulation via CD2 activated multiple signal transduction pathways, resulting in phosphorylation of distinct sites of stathmin, the combination of which only partially overlaps the CD3- and CD28-induced patterns. The partial redundancy of the three T cell activation pathways was evidenced by the phosphorylation of Ser25 and Ser38, substrates of MAP kinases and of the cdk family kinase(s), respectively. Conversely, the phosphorylation of Ser16 of stathmin was observed in response to both CD2 and CD28 triggering, but not CD3 triggering, with a kinetics compatible with the lasting activation of CaM kinase II in response to CD2 triggering. In vitro, Ser16 of recombinant human stathmin was phosphorylated also by purified CaM kinase II, and in vivo, CaM kinase II activity was indeed stimulated in CD2-triggered Jurkat cells. Altogether, our results favor an association of CaM kinase II activity with costimulatory signals of T lymphocyte activation and phosphorylation of stathmin on Ser16.
Collapse
Affiliation(s)
- Sabine le Gouvello
- Institut National de la Santé et de la Recherche Médicale U440, Paris, France
| | - Valérie Manceau
- Institut National de la Santé et de la Recherche Médicale U440, Paris, France
| | - André Sobel
- Institut National de la Santé et de la Recherche Médicale U440, Paris, France
| |
Collapse
|
35
|
Heist EK, Srinivasan M, Schulman H. Phosphorylation at the nuclear localization signal of Ca2+/calmodulin-dependent protein kinase II blocks its nuclear targeting. J Biol Chem 1998; 273:19763-71. [PMID: 9677407 DOI: 10.1074/jbc.273.31.19763] [Citation(s) in RCA: 86] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Translocation of protein kinases with broad substrate specificities between different subcellular compartments by activation of signaling pathways is an established mechanism to direct the activity of these enzymes toward particular substrates. Recently, we identified two isoforms of Ca2+/calmodulin-dependent protein kinase II (CaM kinase II), which are targeted to the nucleus by an alternatively spliced nuclear localization signal (NLS). Here we report that cotransfection with constitutively active mutants of CaM kinase I or CaM kinase IV specifically blocks nuclear targeting of CaM kinase II as a result of phosphorylation of a Ser immediately adjacent to the NLS of CaM kinase II. Both CaM kinase I and CaM kinase IV are able to phosphorylate this Ser residue in vitro, and mutagenesis studies suggest that this phosphorylation is both necessary and sufficient to block nuclear targeting. Furthermore, we provide experimental evidence that introduction of a negatively charged residue at this phosphorylation site reduces binding of the kinase to an NLS receptor in vitro, thus providing a mechanism that may explain the blockade of nuclear targeting that we have observed in situ.
Collapse
Affiliation(s)
- E K Heist
- Department of Neurobiology, Stanford University School of Medicine, Stanford, California 94305-5125, USA
| | | | | |
Collapse
|
36
|
Schmitz AA, Pleschke JM, Kleczkowska HE, Althaus FR, Vergères G. Poly(ADP-ribose) modulates the properties of MARCKS proteins. Biochemistry 1998; 37:9520-7. [PMID: 9649335 DOI: 10.1021/bi973063b] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023]
Abstract
In mammalian cells, the formation of DNA strand breaks is accompanied by synthesis of poly(ADP-ribose). This nucleic acid-like homopolymer may modulate protein functions by covalent and/or noncovalent interactions. Here we show that poly(ADP-ribose) binds strongly to the proteins of the myristoylated alanine-rich C kinase substrate (MARCKS) family, MARCKS and MARCKS-related protein (also MacMARCKS or F52). MARCKS proteins are myristoylated proteins associated with membranes and the actin cytoskeleton. As targets for both protein kinase C (PKC) and calmodulin (CaM), MARCKS proteins are thought to mediate cross-talk between these two signal transduction pathways. Dot blot assays show that poly(ADP-ribose) binds to MARCKS proteins at the highly basic effector domain. Complex formation between MARCKS-related protein and CaM as well as phosphorylation of MARCKS-related protein by the catalytic subunit of PKC are strongly inhibited by equimolar amounts of poly(ADP-ribose), suggesting a high affinity of poly(ADP-ribose) for MARCKS-related protein. Binding of MARCKS-related protein to membranes is also inhibited by poly(ADP-ribose). Finally, poly(ADP-ribose) efficiently reverses the actin-filament bundling activity of a peptide corresponding to the effector domain and inhibits the formation of actin filaments in vitro. Our results suggest that MARCKS proteins and actin could be targets of the poly(ADP-ribose) DNA damage signal pathway.
Collapse
Affiliation(s)
- A A Schmitz
- Department of Biophysical Chemistry, Biozentrum, University of Basel, Switzerland
| | | | | | | | | |
Collapse
|
37
|
McCullough LA, Egan TM, Westfall TC. Neuropeptide Y inhibition of calcium channels in PC-12 pheochromocytoma cells. THE AMERICAN JOURNAL OF PHYSIOLOGY 1998; 274:C1290-7. [PMID: 9612216 DOI: 10.1152/ajpcell.1998.274.5.c1290] [Citation(s) in RCA: 25] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
We previously demonstrated, using rat PC-12 pheochromocytoma cells differentiated to a sympathetic neuronal phenotype with nerve growth factor (NGF), that neuropeptide Y (NPY) inhibits catecholamine synthesis as well as release. Inquiry into the mechanisms of these inhibitions implicated distinct pathways involving reduction of Ca2+ influx through voltage-activated Ca2+ channels. In the present investigation the effects of NPY on whole cell Ba2+ currents were examined to obtain direct evidence supporting the mechanisms suggested by those studies. NPY was found to inhibit the voltage-activated Ba2+ current in NGF-differentiated PC-12 cells in a reversible fashion with an EC50 of 13 nM. This inhibition was pertussis toxin sensitive and resulted from NPY modulation of L- and N-type Ca2+ channels. The inhibition of L-type channels was not seen with < 1 nM free intracellular Ca2+ or when protein kinase C (PKC) was inhibited by chelerythrine or PKC-(19-31). Furthermore, the effect of NPY on L-type channels was mimicked by the PKC activator phorbol 12-myristate 13-acetate. These studies demonstrate that, in addition to inhibition of N-type Ca2+ channels, in NGF-differentiated PC-12 cells NPY inhibits L-type Ca2+ channels via an intracellular Ca(2+)- and PKC-dependent pathway.
Collapse
Affiliation(s)
- L A McCullough
- Department of Pharmacological and Physiological Science, Saint Louis University Health Sciences Center, Missouri 63104, USA
| | | | | |
Collapse
|
38
|
Barria A, Derkach V, Soderling T. Identification of the Ca2+/calmodulin-dependent protein kinase II regulatory phosphorylation site in the alpha-amino-3-hydroxyl-5-methyl-4-isoxazole-propionate-type glutamate receptor. J Biol Chem 1997; 272:32727-30. [PMID: 9407043 DOI: 10.1074/jbc.272.52.32727] [Citation(s) in RCA: 307] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023] Open
Abstract
Ca2+/CaM-dependent protein kinase II (CaM-KII) can phosphorylate and potentiate responses of alpha-amino3-hydroxyl-5-methyl-4-isoxazole-propionate-type glutamate receptors in a number of systems, and recent studies implicate this mechanism in long term potentiation, a cellular model of learning and memory. In this study we have identified this CaM-KII regulatory site using deletion and site-specific mutants of glutamate receptor 1 (GluR1). Only mutations affecting Ser831 altered the 32P peptide maps of GluR1 from HEK-293 cells co-expressing an activated CaM-KII. Likewise, when CaM-KII was infused into cells expressing GluR1, the Ser831 to Ala mutant failed to show potentiation of the GluR1 current. The Ser831 site is specific to GluR1, and CaM-KII did not phosphorylate or potentiate current in cells expressing GluR2, emphasizing the importance of the GluR1 subunit in this regulatory mechanism. Because Ser831 has previously been identified as a protein kinase C phosphorylation site (Roche, K. W., O'Brien, R. J., Mammen, A. L., Bernhardt, J., and Huganir, R. L. (1996) Neuron 16, 1179-1188), this raises the possibility of synergistic interactions between CaM-KII and protein kinase C in regulating synaptic plasticity.
Collapse
Affiliation(s)
- A Barria
- Vollum Institute, Oregon Health Sciences University, Portland, Oregon 97201, USA
| | | | | |
Collapse
|
39
|
Abstract
Amphetamine is taken up through the dopamine transporter in nerve terminals and enhances the release of dopamine. We previously found that incubation of rat striatal synaptosomes increases phosphorylation of the presynaptic neural-specific protein, neuromodulin (Gnegy et al., Mol. Brain Res. 20:289-293, 1993). Using a state-specific antibody, we now demonstrate that incubation of rat striatal synaptosomes with amphetamine increases levels of neuromodulin phosphorylated at ser41, the protein kinase C substrate site. Phosphorylation was maximal at 5 min at 37 degrees C at concentrations from 100 nM to 10 microM amphetamine. The effect of amphetamine on the phosphorylation of synapsin I at a site specifically phosphorylated by Ca2+/calmodulin-dependent protein kinase II (site 3), was examined using a state-specific antibody for site 3-phosphosynapsin I. Incubation with concentrations of amphetamine from 1 to 100 nM increased the level of site 3-phospho-synapsin I at times from 30 sec to 2 min. The effect of amphetamine on synapsin I phosphorylation was blocked by nomifensine. The presence of calcium in the incubating buffer was required for amphetamine to increase the level of site 3-phospho-synapsin I. The amphetamine-mediated increase in the content of phosphoser41-neuromodulin was less sensitive to extrasynaptosomal calcium. The amphetamine-mediated increase in the content of site 3-phospho-synapsin I persisted in the presence of 10 microM okadaic acid and was not significantly altered by D1 or D2 dopamine receptor antagonists. Preincubation of striatal synaptosomes with 10 microM of the protein kinase C inhibitor, Ro-31-8220, blocked the amphetamine-mediated increases in the levels of both phosphoser41-neuromodulin and site 3-phospho-synapsin I. Our results demonstrate that amphetamine can alter phosphorylation-related second messenger activities in the synaptosome.
Collapse
Affiliation(s)
- S Iwata
- Department of Pharmacology, University of Michigan Medical School, Ann Arbor 48109-0632, USA
| | | | | |
Collapse
|
40
|
Raufman JP, Malhotra R, Raffaniello RD. Regulation of calcium-induced exocytosis from gastric chief cells by protein phosphatase-2B (calcineurin). BIOCHIMICA ET BIOPHYSICA ACTA 1997; 1357:73-80. [PMID: 9202177 DOI: 10.1016/s0167-4889(97)00023-2] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
The molecular mechanisms whereby calcium stimulates secretion are uncertain. In the present study, we used streptolysin O (SLO)-permeabilized chief cells from guinea pig stomach to investigate whether protein phosphatase-2B (calcineurin), a calcium/calmodulin-dependent, serine/threonine phosphatase plays a role in mediating calcium-induced pepsinogen secretion. Preincubation of cells with alpha-naphthylphosphate, a non-specific phosphatase inhibitor, decreased calcium-induced secretion. Likewise, specific inhibitors of protein phosphatase-2B (cyclosporin-A and FK-506) caused a dose-dependent reduction in calcium-induced pepsinogen secretion. Moreover, in intact cells, cyclosporin-A and FK-506 inhibited pepsinogen secretion caused by cholecystokinin, carbamylcholine and A23187, agonists known to increase chief cell cytosolic calcium. Okadaic acid, an inhibitor of protein phosphatase-1 and -2A, had no effect on secretion caused by these agonists. Chief cell calcium-dependent phosphatase activity, measured using radiolabeled casein as substrate, was reduced selectively by inhibitors of protein phosphatase-2B. Endogenous substrates for calcium/calmodulin-dependent phosphatase activity were identified by analyzing chief cell lysates using 2-dimensional gel electrophoresis. Increasing the cytosolic calcium concentration resulted in dephosphorylation of a 55-kDa, acidic cytoskeletal protein. FK-506 inhibited dephosphorylation of this protein. Thus, in permeabilized chief cells, specific inhibitors of protein phosphatase-2B inhibit calcium-induced pepsinogen secretion, calcium/calmodulin-dependent phosphatase activity and calcium-induced dephosphorylation of a 55-kDa, acidic cytoskeletal protein. These results support the hypothesis that protein phosphatase-2B (calcineurin) plays an important role in mediating calcium-induced exocytosis.
Collapse
Affiliation(s)
- J P Raufman
- Division of Gastroenterology, University of Arkansas for Medical Sciences, Little Rock 72205-7199, USA
| | | | | |
Collapse
|
41
|
Expression and phosphorylation of a MARCKS-like protein in gastric chief cells: Further evidence for modulation of pepsinogen secretion by interaction of Ca2+/calmodulin with protein kinase C. J Cell Biochem 1997. [DOI: 10.1002/(sici)1097-4644(19970301)64:3<514::aid-jcb18>3.0.co;2-f] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
42
|
Chen SJ, Sweatt JD, Klann E. Enhanced phosphorylation of the postsynaptic protein kinase C substrate RC3/neurogranin during long-term potentiation. Brain Res 1997; 749:181-7. [PMID: 9138717 DOI: 10.1016/s0006-8993(96)01159-6] [Citation(s) in RCA: 61] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Long-term potentiation (LTP) is a sustained strengthening of synaptic connections that occurs in the mammalian hippocampus, and is a cellular mechanism likely to contribute to memory formation. One question of current interest is whether the biochemical mechanisms responsible for the maintenance of LTP have a presynaptic or postsynaptic locus. We have determined that the phosphorylation of the postsynaptic protein kinase (PKC) substrate RC3/neurogranin is increased in the maintenance phase of LTP, and that the induction of this effect is dependent on activation of the N-methyl-D-aspartate (NMDA) subtype of glutamate receptors. The sustained increase in RC3/neurogranin phosphorylation requires ongoing protein kinase activity, as application of the protein kinase inhibitor H-7 after LTP induction can reverse the increased RC3/neurogranin phosphorylation. Overall, these data are evidence for postsynaptic biochemical changes in the maintenance of LTP. They also implicate RC3/neurogranin as a downstream effector of PKC activity in LTP that could contribute to physiologic expression of LTP.
Collapse
Affiliation(s)
- S J Chen
- Division of Neuroscience, Baylor College of Medicine, Houston, TX 77030, USA
| | | | | |
Collapse
|
43
|
Tan SE, Chen SS. The activation of calcium/calmodulin-dependent protein kinase II after glutamate or potassium stimulation in hippocampal slices. Brain Res Bull 1997; 43:269-73. [PMID: 9227836 DOI: 10.1016/s0361-9230(97)00004-x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023]
Abstract
Two forms of long-term potentiation (LTP), N-methyl-D-aspartate receptor (NMDAR) dependent and non-NMDAR dependent, have been reported in hippocampal CA1 and CA3, respectively. The present study examined the activation of CaM-kinase II (calcium/calmodulin-dependent protein kinase II) in CA1 and CA3 areas after glutamate or potassium stimulation. Rat hippocampal slices were preincubated with one of the drugs (EGTA, DL-APV, CNQX, AP3, nitrendipine, KN-62, staurosporin, and H-89) before they were stimulated with either glutamate/glycine (100 microM/1 microM) or KCl (60 mM). Hippocampal CA1 area and CA3 area were then dissected and CaM-kinase II activities were assayed in vitro. Glutamate and KCl stimulations enhanced the percentage of Ca(2+)-independent CaM-kinase II activity in CA1 area. This enhancement was suppressed by EGTA, DL-APV, CNQX, or KN-62, suggesting that the neuronal stimulation effect in CA1 area was mediating through NMDA receptors. Conversely, there was no significant enhancement of CaM-kinase II activity in the CA3 area after glutamate or KCl stimulation. Nevertheless, the percentage of calcium-independent CaM-kinase II activity in the CA3 area was suppressed by EGTA, nitrendipine, KN-62, staurosporin, or H-89, indicating that the activity of CaM-kinase II in the CA3 area was independent of NMDA receptor activation.
Collapse
Affiliation(s)
- S E Tan
- Department of Psychology, Kaohsiung Medical College, Taiwan, R.O.C
| | | |
Collapse
|
44
|
Zhao Y, Davis HW. Thrombin-induced phosphorylation of the myristoylated alanine-rich C kinase substrate (MARCKS) protein in bovine pulmonary artery endothelial cells. J Cell Physiol 1996; 169:350-7. [PMID: 8908202 DOI: 10.1002/(sici)1097-4652(199611)169:2<350::aid-jcp14>3.0.co;2-d] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Myristoylated alanine-rich C kinase substrates (MARCKS) is a prominent protein kinase C (PKC) substrate that is targeted to the plasma membrane by an aminoterminal myristoyl group. In its nonphosphorylated form, MARCKS cross-links Factin and binds calmodulin (CaM) reciprocally. However, upon phosphorylation by PKC, MARCKS release the actin or CaM MARCKS may therefore act as a CaM sink in resting cells and regulate CaM availability during cell activation. We have demonstrated previously that thrombin-induced myosin light chain (MLC) phosphorylation and increased monolayer permeability in bovine pulmonary artery endothelial cells (BPAEC) require both PKC-and CaM-dependent pathways. We therefore decided to investigate the phosphorylation of MARCKS in BPAEC to ascertain whether this occurs in a temporally relevant manner to participate in the thrombin-induced events. MARCKS is phosphorylated in response to thrombin with a time course similar to that seen with MLC. As expected, MARCKS is also phosphorylated by phorbol 12-myristate 13 acetate (PMA), a PKC activator, but with a slower onset and more prolonged duration. Bradykinin also enhances MARCKS phosphorylation in BPAEC, but histamine does not. MARCKS is distributed evently between the membrane and cytosol in BPAEC, and neither thrombin nor PMA caused significant translocation of the protein. Specific PKC inhibitors attenuated MARCKS phosphorylation by either thrombin or PMA. Since thrombin-induced MLC phosphorylation is also attenuated by these inhibitors, MARCKS may be involved in MLC kinase activation and subsequent BPAEC contraction. W7, a CaM antagonist, enhances the phosphorylation of MARCKS. This was expected since CaM binding to MARCKS has been shown to decrease MARCKS phosphorylation by PKC. On the other hand, tyrosine kinase inhibitors, genistein and tyrphostin, attenuate MARCKS phosphorylation but have no effect on MLC phosphorylation, suggesting that MARCKS may be phosphorylated by kinases other than PKC. Phosphorylation of MARCKS outside the PKC phosphorylation domain would not be expected to induce the release of CaM. These data provide support for the hypothesis that MARCKS may serve as a regulator of CaM availability in BPAEC.
Collapse
Affiliation(s)
- Y Zhao
- Department of Internal Medicine (Pulmonary & Critical Care Medicine), University of Cincinnati Medical Center, Ohio 45267-0564, USA
| | | |
Collapse
|
45
|
Ueno E, Rosenberg P. Differential effects of snake venom phospholipase A2 neurotoxin (beta-bungarotoxin) and enzyme (Naja naja atra) on protein kinases. Biochem Pharmacol 1996; 52:1287-93. [PMID: 8937437 DOI: 10.1016/0006-2952(96)00484-4] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
The phospholipase A2 (PLA2) neurotoxin, beta-bungarotoxin (beta-BuTX), presynaptically alters acetylcholine release. We previously found that beta-BuTX inhibits protein phosphorylation in rat brain synaptosomes. This inhibition was not due to the inhibition of ATP synthesis, the action of arachidonic acid (AA) metabolites, or the stimulation of phosphatase activities. A typical PLA2 enzyme from Naja naja atra (N. n. atra) venom also inhibited phosphorylation but with lesser potency than that of beta-BuTX. We now report the effects of beta-BuTX and N. n. atra PLA2 on the activities of protein kinases. Treatments of synaptic plasma membrane or cytosol with N. n. atra PLA2 stimulated the activities of cAMP-dependent kinase, Ca2+/calmodulin-dependent kinase II, and protein kinase C (PKC), whereas beta-BuTX had no effect on these kinases. Calyculin A, a phosphatase-1 and -2A inhibitor, increased the stimulation of phosphorylation by N. n. atra PLA2, indicating that the stimulation is not due to an inhibition of phosphatase activities. The stimulation of PKC by N. n. atra PLA2 appears to be mediated by free fatty acids (FFAs) resulting from phospholipid hydrolysis by PLA2, since (1) treatment of either synaptic plasma membrane or cytosol with N. n. atra PLA2 produced large amounts of FFAs, and (2) AA, an exogenous FFA, stimulated PKC activity to an extent similar to that caused by N. n. atra PLA2. Thus, the mechanisms of action of beta-BuTX and N. n. atra PLA2 appear quite different from each other although both agents inhibit phosphorylation in intact synaptosomes.
Collapse
Affiliation(s)
- E Ueno
- Department of Pharmaceutical Sciences, University of Connecticut, School of Pharmacy, Storrs 06269, USA
| | | |
Collapse
|
46
|
Schleiff E, Schmitz A, McIlhinney RA, Manenti S, Vergères G. Myristoylation does not modulate the properties of MARCKS-related protein (MRP) in solution. J Biol Chem 1996; 271:26794-802. [PMID: 8900160 DOI: 10.1074/jbc.271.43.26794] [Citation(s) in RCA: 39] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/02/2023] Open
Abstract
The members of the myristoylated alanine-rich C kinase substrate (MARCKS) family are proteins essential for brain development and phagocytosis. MARCKS proteins bind to actin filaments and calmodulin (CaM) and are phosphorylated by protein kinase C. In order to investigate how these interactions are regulated, we have characterized the properties of both the myristoylated (myr) and unmyristoylated (unmyr) forms of recombinant MARCKS-related protein (MRP), a 20-kDa member of the MARCKS family. Ultracentrifugation and circular dichroic spectroscopy reveal that MRP is an elongated protein, with an axis ratio estimated between 7 and 12 and with an apparent random coil conformation. MRP binds to CaM with high affinity (Kd,myr = 4 nM; Kd,unmyr = 7 nM) and with a second order rate constant, k+1,unmyr, of 1.6 x 10(8) M-1 s-1. In contrast to classical ligands such as the myosin light chain kinase, binding of MRP to CaM does not induce the formation of an alpha-helix in MRP. The catalytic subunit of protein kinase C (PKM) phosphorylates myr MRP with high affinity ([S]0.5 = 3.5 microM), positive cooperativity (nH = 2.5) and a turnover number of 130 min-1. CaM inhibits the phosphorylation of myr MRP with a half-maximum rate of phosphorylation at a [CaM]/[MRP] ratio of 0.7, indicating that CaM might efficiently regulate the phosphorylation of MRP in vivo. Interestingly, Ca2+ inhibits the binding of MRP to CaM as well as its phosphorylation by PKM in the millimolar concentration range, suggesting that MRP has a weak affinity for Ca2+. Finally, unmyr MRP can be stoichiometrically myristoylated by N-myristoyl transferase in vitro. Since neither binding of CaM nor phosphorylation by PKM inhibits myristoylation, the N terminus of unmyr MRP is exposed on the surface of the protein and is well separated from the effector domain. In view of the observations that unmyr and myr MRP do not exhibit significant differences in their properties in solution, the function of myristoylation is most probably to modulate the interactions of MRP with membranes.
Collapse
Affiliation(s)
- E Schleiff
- Department of Biophysical Chemistry, Biozentrum, University of Basel, Klingelbergstrasse 70, 4056 Basel, Switzerland
| | | | | | | | | |
Collapse
|
47
|
Abstract
The observation that autophosphorylation converts CaM kinase II from the Ca(2+)-dependent form to the Ca(2+)-independent form has led to speculation that the formation of the Ca(2+)-independent form of the enzyme could encode frequency of synaptic usage and serve as a molecular explanation of "memory". In cultured rat hippocampal neurons, glutamate elevated the Ca(2+)-independent activity of CaM kinase II through autophosphorylation, and this response was blocked by an NMDA receptor antagonist, D-2-amino-5-phosphonopentanoate (AP5). In addition, we confirmed that high, but not low frequency stimulation, applied to two groups of CA1 afferents in the rat hippocampus, resulted in LTP induction with concomitant long-lasting increases in Ca(2+)-independent and total activities of CaM kinase II. In experiments with 32P-labeled hippocampal slices, the LTP induction in the CA1 region was associated with increases in autophosphorylation of both alpha and beta subunits of CaM kinase II 1 h after LTP induction. Significant increases in phosphorylation of endogenous CaM kinase II substrates, synapsin I and microtubule-associated protein 2 (MAP2), which are originally located in presynaptic and postsynaptic regions, respectively, were also observed in the same slice. All these changes were prevented when high frequency stimulation was applied in the presence of AP5 or a calmodulin antagonist, calmidazolium. Furthermore, in vitro phosphorylation of the AMPA receptor by CaM kinase II was reported in the postsynaptic density and infusion of the constitutively active CaM kinase II into the hippocampal neurons enhanced kainate-induced response. These results support the idea that CaM kinase II contributes to the induction of hippocampal LTP in both postsynaptic and presynaptic regions through phosphorylation of target proteins such as the AMPA receptor, MAP2 and synapsin I.
Collapse
Affiliation(s)
- K Fukunaga
- Department of Pharmacology, Kumamoto University School of Medicine, Japan
| | | | | |
Collapse
|
48
|
Williams CL, Phelps SH, Porter RA. Expression of Ca2+/calmodulin-dependent protein kinase types II and IV, and reduced DNA synthesis due to the Ca2+/calmodulin-dependent protein kinase inhibitor KN-62 (1-[N,O-bis(5-isoquinolinesulfonyl)-N-methyl-L-tyrosyl]-4-phenyl piperazine) in small cell lung carcinoma. Biochem Pharmacol 1996; 51:707-15. [PMID: 8615909 DOI: 10.1016/s0006-2952(95)02393-3] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Because changes in intracellular Ca2+ affect progression through the mitotic cell cycle, we investigated the role of Ca2+-binding proteins in regulating cell cycle progression. Evidence was found demonstrating that the activation of Ca2+/calmodulin-dependent protein kinase (CaM kinase) inhibits cell cycle progression in small cell lung carcinoma (SCLC) cells. We also demonstrated that SCLC cells express both CaM kinase type II (CaMKII) and CaM kinase type IV (CaMKIV). Five independent SCLC cell lines expressed proteins reactive with antibody to the CaMKII beta subunit, but none expressed detectable proteins reactive with antibody to the CaMKII alpha subunit. All SCLC cell lines tested expressed both the alpha and beta isoforms of CaMKIV. Immunoprecipitation of CaMKII from SCLC cells yielded multiple proteins that autophosphorylated in the presence of Ca2+ / calmodulin. Autophosphorylation was inhibited by the CaMKII(281-302) peptide, which corresponds to the CaMKII autoinhibitory domain, and by 1-[N,O-bis(5-isoquinolinesulfonyl)-N-methyl-L-tyrosyl]-4- phenylpiperazine (KN-62), a specific CaM kinase antagonist. Influx of Ca2+ through voltage-gated Ca2+ channels stimulated phosphorylation of CaMKII in SCLC cells, and this was inhibited by KN-62. Incubation of SCLC cells of KN-62 potently inhibited DNA synthesis, and slowed progression through S phase. Similar anti-proliferative effects of KN-62 occurred in SK-N-SH human neuroblastoma cells, which express both CaMKII and CaMKIV, and in K562 human chronic myelogenous leukemia cells, which express CaMKII but not CaMKIV. The expression of both CaMKII and CaMKIV by SCLC cells, and the sensitivity of these cells to the anti-proliferative effects of KN-62, suggest a role for CaM kinase in regulating SCLC proliferation.
Collapse
Affiliation(s)
- C L Williams
- Molecular Pharmacology Laboratory, Guthrie Research Institute, Sayre, PA 18840, USA
| | | | | |
Collapse
|
49
|
Abraham ST, Benscoter H, Schworer CM, Singer HA. In situ Ca2+ dependence for activation of Ca2+/calmodulin-dependent protein kinase II in vascular smooth muscle cells. J Biol Chem 1996; 271:2506-13. [PMID: 8576214 DOI: 10.1074/jbc.271.5.2506] [Citation(s) in RCA: 52] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
Activation of Ca2+/calmodulin (CaM)-dependent protein kinase II (CaM kinase II) and development of the Ca2+/CaM-independent (autonomous) form of the kinase was investigated in cultured vascular smooth muscle (VSM) cells. Within 15 s of ionomycin (1 microM) exposure 52.7 +/- 4.4% of the kinase became autonomous, a response that was partially maintained for at least 10 min. This correlated with 32P phosphorylation of CaM kinase II delta-subunits in situ and was abolished by pretreatment with the CaM kinase II inhibitor KN-93. The in situ Ca2+ dependence for generating autonomous CaM kinase II was determined in cells selectively permeabilized to Ca2+ and depleted of sarcoplasmic reticulum Ca2+ by pretreatment with thapsigargin. Analysis of the resulting curve revealed an EC50 (concentration producing 50% of maximal response) of 692 +/- 28 nM [Ca2+]i, a maximum of 68 +/- 2% of the total activity becoming autonomous reflecting nearly complete activation of CaM kinase II and a Hill slope of 3, indicating a highly cooperative process. Based on this dependence and measured [Ca2+]i responses in intact cells, increases in autonomous activity stimulated by angiotensin II, vasopressin and platelet-derived growth factor-BB (4.6-, 2-, and 1.7-fold, respectively) were unexpectedly high. In intact cells stimulated by ionomycin, the correlation between autonomous activity and [Ca2+]i resulted in a parallel curve with an EC50 of 304 +/- 23 nM [Ca2+]i. This apparent increase in Ca2+ sensitivity for generating autonomous activity in intact VSM cells was eliminated by thapsigargin pretreatment. We conclude that alteration of [Ca2+]i over a physiological range activates CaM kinase II in VSM and that this process is facilitated by release of Ca2+ from intracellular pools which initiates cooperative autophosphorylation and consequent generation of autonomous CaM kinase II activity.
Collapse
Affiliation(s)
- S T Abraham
- Sigfried and Janet Weis Center for Research, Geisinger Clinic, Danville, Pennsylvania 17822, USA
| | | | | | | |
Collapse
|
50
|
Chang S, Hemmings HC, Aderem A. Stimulus-dependent phosphorylation of MacMARCKS, a protein kinase C substrate, in nerve termini and PC12 cells. J Biol Chem 1996; 271:1174-8. [PMID: 8557647 DOI: 10.1074/jbc.271.2.1174] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023] Open
Abstract
MacMARCKS (also known as myristoylated alanine-rich C kinase substrate (MARCKS)-related protein) is a member of the MARCKS family of protein kinase C substrates, which binds Ca2+/calmodulin in a phosphorylation-dependent manner. Immunoprecipitation demonstrated that MacMARCKS is present in both PC12 cells and in neurons. Upon depolarization of PC12 cells with 60 mM KCl, MacMARCKS phosphorylation increased 4-fold over basal levels in a Ca(2+)-dependent manner. By immunofluorescence microscopy, MacMARCKS was colocalized in PC12 cells to neurite tips with the synaptic vesicle membrane protein synaptophysin and to vesicles in the perinuclear region. Subcellular fractionation demonstrated that MacMARCKS associates tightly with membranes in PC12 cells. In Percoll-purified rat cerebrocortical synaptosomes, depolarization with 60 mM KCl in the presence of exogenous Ca2+ transiently increased MacMARCKS phosphorylation, whereas phorbol ester promoted a sustained increase in MacMARCKS phosphorylation. Subcellular fractionation of rat brain indicated that MacMARCKS was present in both soluble and particulate fractions; particulate MacMARCKS was associated with both small vesicles and highly purified synaptic vesicles. These results are consistent with a role for MacMARCKS in integrating Ca(2+)-calmodulin and protein kinase C-dependent signals in the regulation of neurosecretion.
Collapse
Affiliation(s)
- S Chang
- Laboratory of Signal Transduction, Rockefeller University, New York, New York 10021-6399, USA
| | | | | |
Collapse
|