1
|
Ishii T, Kajimoto T, Kikkawa S, Narasaki S, Noguchi S, Imamura S, Harada K, Hide I, Tanaka S, Tsutsumi YM, Sakai N. Protein kinase C (PKC) inhibitor Calphostin C activates PKC in a light-dependent manner at high concentrations via the production of singlet oxygen. Eur J Pharmacol 2024; 984:177036. [PMID: 39368603 DOI: 10.1016/j.ejphar.2024.177036] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/28/2024] [Revised: 07/20/2024] [Accepted: 10/03/2024] [Indexed: 10/07/2024]
Abstract
Calphostin C (Cal-C) is a protein kinase C (PKC) inhibitor that binds to its C1 domain. The aim of the present study was to elucidate the action of Cal-C in addition to PKC inhibition. First, we confirmed that Cal-C at low concentrations (<200 nM) inhibit phorbol ester-induced PKC translocation and G-protein-coupled receptor (GPCR)-mediated PKC activation. Cal-C at higher concentrations (>2 μM) increased intracellular calcium ion concentrations ([Ca2+]i) in a concentration-dependent manner. The origin of this increase is the mobilization of the endoplasmic reticulum (ER), which does not involve GPCR or ryanodine receptors. Cal-C at high concentrations also cause structural changes in the ER, such as the formation of vacuoles and aggregates, and calcium leakage from the ER. At 2 μM, Cal-C translocated a calcium-sensitive PKCα. Studies using a C-kinase activity reporter and a myristoylated alanine-rich protein kinase C substrate fused with green fluorescent protein (GFP) have also revealed that Cal-C at high concentrations activate PKC in living cells. Additionally, the PKC-activating effects of Cal-C were light-dependent. Finally, studies using Si-DMA, an indicator of singlet oxygen, showed that Cal-C at high concentrations generated singlet oxygen, causing structural changes in the ER and leakage of calcium into the cytosol, which triggered PKC activation. This study confirms the novel action of Cal-C, solely considered a PKC inhibitor. Cal-C acted as a PKC inhibitor at low concentrations and a PKC activator at high concentrations by generating singlet oxygen in a light-dependent manner, suggesting that Cal-C can be used in photodynamic therapy.
Collapse
Affiliation(s)
- Tomomi Ishii
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical & Health Sciences, Hiroshima University, Japan; Department of Anesthesiology and Critical Care, Graduate School of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Taketoshi Kajimoto
- Division of Biochemistry, Department of Biochemistry and Molecular Biology, Kobe University Graduate School of Medicine, Japan
| | - Satoshi Kikkawa
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Soshi Narasaki
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical & Health Sciences, Hiroshima University, Japan; Department of Anesthesiology and Critical Care, Graduate School of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Soma Noguchi
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Serika Imamura
- Department of Dental Anesthesiology, Graduate School of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Kana Harada
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Izumi Hide
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Shigeru Tanaka
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Yasuo M Tsutsumi
- Department of Anesthesiology and Critical Care, Graduate School of Biomedical & Health Sciences, Hiroshima University, Japan
| | - Norio Sakai
- Department of Molecular and Pharmacological Neuroscience, Graduate School of Biomedical & Health Sciences, Hiroshima University, Japan.
| |
Collapse
|
2
|
Fouedji C, Etémé AS, Tabi CB, Fouda HPE, Kofané TC. Multisolitons-like patterns in a one-dimensional MARCKS protein cyclic model. J Theor Biol 2024; 579:111702. [PMID: 38096977 DOI: 10.1016/j.jtbi.2023.111702] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2023] [Revised: 11/16/2023] [Accepted: 12/08/2023] [Indexed: 12/24/2023]
Abstract
In this paper, we study the nonlinear dynamics of the MARCKS protein between cytosol and cytoplasmic membrane through the modulational instability phenomenon. The reaction-diffusion generic model used here is firstly transformed into a cubic complex Ginzburg-Landau equation. Then, modulational instability (MI) is carried out in order to derive the MI criteria. We find the domains of some parameter space where nonlinear patterns are expected in the model. The analytical results on the MI growth rate predict that phosphorylation and binding rates affect MARCKS dynamics in opposite way: while the phosphorylation rate tends to support highly localized structures of MARCKS, the binding rate in turn tends to slow down such features. On the other hand, self-diffusion process always amplifies the MI phenomenon. These predictions are confirmed by numerical simulations. As a result, the cyclic transport of MARCKS protein from membrane to cytosol may be done by means of multisolitons-like patterns.
Collapse
Affiliation(s)
- Chenceline Fouedji
- Laboratory of Biophysics, Department of Physics, Faculty of Science, University of Yaounde I, P.O. Box 812, Yaounde, Cameroon.
| | - Armand Sylvin Etémé
- Laboratory of Biophysics, Department of Physics, Faculty of Science, University of Yaounde I, P.O. Box 812, Yaounde, Cameroon.
| | - Conrad Bertrand Tabi
- Botswana International University of Science and Technology, P/Bag 16 Palapye, Botswana.
| | - Henri Paul Ekobena Fouda
- Laboratory of Biophysics, Department of Physics, Faculty of Science, University of Yaounde I, P.O. Box 812, Yaounde, Cameroon.
| | - Timoléon Crépin Kofané
- Laboratory of Mechanics, Department of Physics, Faculty of Science, University of Yaounde I, P.O. Box 812, Yaounde, Cameroon.
| |
Collapse
|
3
|
Sasaki S, Takahashi R, Luo Y, Chujo K, Sera T, Kudo S. Spatiotemporal distribution of PKCα, Cdc42, and Rac1 before directed cell migration. Biochem Biophys Res Commun 2021; 584:26-31. [PMID: 34753065 DOI: 10.1016/j.bbrc.2021.10.080] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2021] [Revised: 10/30/2021] [Accepted: 10/31/2021] [Indexed: 11/19/2022]
Abstract
Cdc42 is a key factor in directed cell migration and accumulates at the leading edge of migrating cells. However, what kind of proteins control Cdc42 and when is unclear. After mechanical wounding, protein kinase C α (PKCα), a conventional PKC isozyme, begins to accumulate at the edges of cells adjacent to the wounded cells (WCs). In this study, we hypothesized that PKCα may be implicated in directed cell migration at an early stage before Cdc42 controls the migration. We focused on the spatiotemporal distribution of PKCα, Cdc42, and Rac1 before cell migration. After wounding, at the edges of cells adjacent to the WCs, PKCα accumulation, Cdc42 accumulation, Rac1 accumulation, and filopodia formation occurred in that order. The PKCα inhibitor suppressed Cdc42 accumulation at the cell edges. These results suggest that inhibition of PKCα activity inhibits cell migration. In addition, it is not Cdc42 but PKCα that may decide the direction of cell migration.
Collapse
Affiliation(s)
- Saori Sasaki
- Department of Mechanical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| | - Ryu Takahashi
- Department of Mechanical Engineering, Graduate School of Engineering, Kyushu University, Fukuoka, Japan
| | - Yangfeng Luo
- Graduate School of Systems Life Sciences, Kyushu University, Fukuoka, Japan
| | - Kengo Chujo
- Department of Mechanical Engineering, Graduate School of Engineering, Kyushu University, Fukuoka, Japan
| | - Toshihiro Sera
- Department of Mechanical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan
| | - Susumu Kudo
- Department of Mechanical Engineering, Faculty of Engineering, Kyushu University, Fukuoka, Japan.
| |
Collapse
|
4
|
Iyer DN, Faruq O, Zhang L, Rastgoo N, Liu A, Chang H. Pathophysiological roles of myristoylated alanine-rich C-kinase substrate (MARCKS) in hematological malignancies. Biomark Res 2021; 9:34. [PMID: 33958003 PMCID: PMC8101130 DOI: 10.1186/s40364-021-00286-9] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2021] [Accepted: 04/16/2021] [Indexed: 12/17/2022] Open
Abstract
The myristoylated alanine-rich C-kinase substrate (MARCKS) protein has been at the crossroads of multiple signaling pathways that govern several critical operations in normal and malignant cellular physiology. Functioning as a target of protein kinase C, MARCKS shuttles between the phosphorylated cytosolic form and the unphosphorylated plasma membrane-bound states whilst regulating several molecular partners including, but not limited to calmodulin, actin, phosphatidylinositol-4,5-bisphosphate, and phosphoinositide-3-kinase. As a result of these interactions, MARCKS directly or indirectly modulates a host of cellular functions, primarily including cytoskeletal reorganization, membrane trafficking, cell secretion, inflammatory response, cell migration, and mitosis. Recent evidence indicates that dysregulated expression of MARCKS is associated with the development and progression of hematological cancers. While it is understood that MARCKS impacts the overall carcinogenesis as well as plays a part in determining the disease outcome in blood cancers, we are still at an early stage of interpreting the pathophysiological roles of MARCKS in neoplastic disease. The situation is further complicated by contradictory reports regarding the role of phosphorylated versus an unphosphorylated form of MARCKS as an oncogene versus tumor suppressor in blood cancers. In this review, we will investigate the current body of knowledge and evolving concepts of the physical properties, molecular network, functional attributes, and the likely pathogenic roles of MARCKS in hematological malignancies. Key emphasis will also be laid upon understanding the novel mechanisms by which MARCKS determines the overall disease prognosis by playing a vital role in the induction of therapeutic resistance. Additionally, we will highlight the importance of MARCKS as a valuable therapeutic target in blood cancers and will discuss the potential of existing strategies available to tackle MARCKS-driven blood cancers.
Collapse
Affiliation(s)
- Deepak Narayanan Iyer
- Laboratory medicine program, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada
| | - Omar Faruq
- Laboratory medicine program, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada
| | - Lun Zhang
- Laboratory medicine program, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada
| | - Nasrin Rastgoo
- Laboratory medicine program, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada
| | - Aijun Liu
- Department of Hematology, Beijing Chaoyang Hospital, Capital University, Beijing, China.
| | - Hong Chang
- Laboratory medicine program, Toronto General Hospital, University Health Network, University of Toronto, Toronto, Canada.
| |
Collapse
|
5
|
Sheats MK, Yin Q, Fang S, Park J, Crews AL, Parikh I, Dickson B, Adler KB. MARCKS and Lung Disease. Am J Respir Cell Mol Biol 2019; 60:16-27. [PMID: 30339463 DOI: 10.1165/rcmb.2018-0285tr] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
MARCKS (myristoylated alanine-rich C kinase substrate) is a prominent PKC substrate expressed in all eukaryotic cells. It is known to bind to and cross-link actin filaments, to serve as a bridge between Ca2+/calmodulin and PKC signaling, and to sequester the signaling molecule phosphatidylinositol 4,5-bisphosphate in the plasma membrane. Since the mid-1980s, this evolutionarily conserved and ubiquitously expressed protein has been associated with regulating cellular events that require dynamic actin reorganization, including cellular adhesion, migration, and exocytosis. More recently, translational studies have implicated MARCKS in the pathophysiology of a number of airway diseases, including chronic obstructive pulmonary disease, asthma, lung cancer, and acute lung injury/acute respiratory distress syndrome. This article summarizes the structure and cellular function of MARCKS (also including MARCKS family proteins and MARCKSL1 [MARCKS-like protein 1]). Evidence for MARCKS's role in several lung diseases is discussed, as are the technological innovations that took MARCKS-targeting strategies from theoretical to therapeutic. Descriptions and updates derived from ongoing clinical trials that are investigating inhalation of a MARCKS-targeting peptide as therapy for patients with chronic bronchitis, lung cancer, and ARDS are provided.
Collapse
Affiliation(s)
| | - Qi Yin
- 2 Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina; and
| | - Shijing Fang
- 2 Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina; and
| | - Joungjoa Park
- 2 Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina; and
| | - Anne L Crews
- 2 Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina; and
| | - Indu Parikh
- 3 BioMarck Pharmaceuticals, Durham, North Carolina
| | | | - Kenneth B Adler
- 2 Department of Molecular Biomedical Sciences, North Carolina State University College of Veterinary Medicine, Raleigh, North Carolina; and
| |
Collapse
|
6
|
Hashimoto T, Mogami H, Tsuriya D, Morita H, Sasaki S, Kumada T, Suzuki Y, Urano T, Oki Y, Suda T. G-protein-coupled receptor 40 agonist GW9508 potentiates glucose-stimulated insulin secretion through activation of protein kinase Cα and ε in INS-1 cells. PLoS One 2019; 14:e0222179. [PMID: 31498851 PMCID: PMC6733457 DOI: 10.1371/journal.pone.0222179] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/26/2018] [Accepted: 08/23/2019] [Indexed: 11/18/2022] Open
Abstract
OBJECTIVE The mechanism by which G-protein-coupled receptor 40 (GPR40) signaling amplifies glucose-stimulated insulin secretion through activation of protein kinase C (PKC) is unknown. We examined whether a GPR40 agonist, GW9508, could stimulate conventional and novel isoforms of PKC at two glucose concentrations (3 mM and 20 mM) in INS-1D cells. METHODS Using epifluorescence microscopy, we monitored relative changes in the cytosolic fluorescence intensity of Fura2 as a marker of change in intracellular Ca2+ ([Ca2+]i) and relative increases in green fluorescent protein (GFP)-tagged myristoylated alanine-rich C kinase substrate (MARCKS-GFP) as a marker of PKC activation in response to GW9508 at 3 mM and 20 mM glucose. To assess the activation of the two PKC isoforms, relative increases in membrane fluorescence intensity of PKCα-GFP and PKCε-GFP were measured by total internal reflection fluorescence microscopy. Specific inhibitors of each PKC isotype were constructed and synthesized as peptide fusions with the third α-helix of the homeodomain of Antennapedia. RESULTS At 3 mM glucose, GW9508 induced sustained MARCKS-GFP translocation to the cytosol, irrespective of changes in [Ca2+]i. At 20 mM glucose, GW9508 induced sustained MARCKS-GFP translocation but also transient translocation that followed sharp increases in [Ca2+]i. Although PKCα translocation was rarely observed, PKCε translocation to the plasma membrane was sustained by GW9508 at 3 mM glucose. At 20 mM glucose, GW9508 induced transient translocation of PKCα and sustained translocation as well as transient translocation of PKCε. While the inhibitors (75 μM) of each PKC isotype reduced GW9508-potentiated, glucose-stimulated insulin secretion in INS-1D cells, the PKCε inhibitor had a more potent effect. CONCLUSION GW9508 activated PKCε but not PKCα at a substimulatory concentration of glucose. Both PKC isotypes were activated at a stimulatory concentration of glucose and contributed to glucose-stimulated insulin secretion in insulin-producing cells.
Collapse
Affiliation(s)
- Takuya Hashimoto
- 2nd Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
- * E-mail:
| | - Hideo Mogami
- Department of Health and Nutrition, Tokoha University, Shizuoka, Japan
| | - Daisuke Tsuriya
- 2nd Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Hiroshi Morita
- 2nd Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Shigekazu Sasaki
- 2nd Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Tatsuro Kumada
- Department of Occupational Therapy, Tokoha University, Shizuoka, Japan
| | - Yuko Suzuki
- Department of Medical Physiology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Tetsumei Urano
- Department of Medical Physiology, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Yutaka Oki
- 2nd Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
- Department of Family and Community Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
| | - Takafumi Suda
- 2nd Department of Internal Medicine, Hamamatsu University School of Medicine, Shizuoka, Japan
| |
Collapse
|
7
|
Li N, Qian S, Li B, Zhan X. Quantitative analysis of the human ovarian carcinoma mitochondrial phosphoproteome. Aging (Albany NY) 2019; 11:6449-6468. [PMID: 31442208 PMCID: PMC6738437 DOI: 10.18632/aging.102199] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/15/2019] [Accepted: 08/10/2019] [Indexed: 05/02/2023]
Abstract
To investigate the existence and their potential biological roles of mitochondrial phosphoproteins (mtPPs) in human ovarian carcinoma (OC), mitochondria purified from OC and control tissues were analyzed with TiO2 enrichment-based iTRAQ quantitative proteomics. Totally 67 mtPPs with 124 phosphorylation sites were identified, which of them included 48 differential mtPPs (mtDPPs). Eighteen mtPPs were reported previously in OCs, and they were consistent in this study compared to previous literature. GO analysis revealed those mtPPs were involved in multiple cellular processes. PPI network indicated that those mtPPs were correlated mutually, and some mtPPs acted as hub molecules, such as EIF2S2, RPLP0, RPLP2, CFL1, MYH10, HSP90, HSPD1, PSMA3, TMX1, VDAC2, VDAC3, TOMM22, and TOMM20. Totally 32 mtPP-pathway systems (p<0.05) were enriched and clustered into 15 groups, including mitophagy, apoptosis, deubiquitination, signaling by VEGF, RHO-GTPase effectors, mitochondrial protein import, translation initiation, RNA transport, cellular responses to stress, and c-MYC transcriptional activation. Totally 29 mtPPs contained a certain protein domains. Upstream regulation analysis showed that TP53, TGFB1, dexamethasone, and thapsigargin might act as inhibitors, and L-dopa and forskolin might act as activators. This study provided novel insights into mitochondrial protein phosphorylations and their potential roles in OC pathogenesis and offered new biomarker resource for OCs.
Collapse
Affiliation(s)
- Na Li
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China
| | - Shehua Qian
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China
| | - Biao Li
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China
| | - Xianquan Zhan
- Key Laboratory of Cancer Proteomics of Chinese Ministry of Health, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China
- Hunan Engineering Laboratory for Structural Biology and Drug Design, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China
- State Local Joint Engineering Laboratory for Anticancer Drugs, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, Hunan, P. R. China
| |
Collapse
|
8
|
Wang J, Ji X, Liu J, Zhang X. Serine/Threonine Protein Kinase STK16. Int J Mol Sci 2019; 20:ijms20071760. [PMID: 30974739 PMCID: PMC6480182 DOI: 10.3390/ijms20071760] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/25/2019] [Revised: 04/05/2019] [Accepted: 04/08/2019] [Indexed: 12/18/2022] Open
Abstract
STK16 (Ser/Thr kinase 16, also known as Krct/PKL12/MPSK1/TSF-1) is a myristoylated and palmitoylated Ser/Thr protein kinase that is ubiquitously expressed and conserved among all eukaryotes. STK16 is distantly related to the other kinases and belongs to the NAK kinase family that has an atypical activation loop architecture. As a membrane-associated protein that is primarily localized to the Golgi, STK16 has been shown to participate in the TGF-β signaling pathway, TGN protein secretion and sorting, as well as cell cycle and Golgi assembly regulation. This review aims to provide a comprehensive summary of the progress made in recent research about STK16, ranging from its distribution, molecular characterization, post-translational modification (fatty acylation and phosphorylation), interactors (GlcNAcK/DRG1/MAL2/Actin/WDR1), and related functions. As a relatively underexplored kinase, more studies are encouraged to unravel its regulation mechanisms and cellular functions.
Collapse
Affiliation(s)
- Junjun Wang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China.
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei 230026, China.
| | - Xinmiao Ji
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China.
| | - Juanjuan Liu
- School of Life Sciences, Anhui University, Hefei 230601, China.
| | - Xin Zhang
- High Magnetic Field Laboratory, Key Laboratory of High Magnetic Field and Ion Beam Physical Biology, Hefei Institutes of Physical Science, Chinese Academy of Sciences, Hefei 230031, China.
- Science Island Branch of Graduate School, University of Science and Technology of China, Hefei 230026, China.
- Institutes of Physical Science and Information Technology, Anhui University, Hefei 230601, China.
| |
Collapse
|
9
|
Polyphosphoinositides in the nucleus: Roadmap of their effectors and mechanisms of interaction. Adv Biol Regul 2019; 72:7-21. [PMID: 31003946 DOI: 10.1016/j.jbior.2019.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/20/2019] [Revised: 04/02/2019] [Accepted: 04/03/2019] [Indexed: 01/01/2023]
Abstract
Biomolecular interactions between proteins and polyphosphoinositides (PPIn) are essential in the regulation of the vast majority of cellular processes. Consequently, alteration of these interactions is implicated in the development of many diseases. PPIn are phosphorylated derivatives of phosphatidylinositol and consist of seven species with different phosphate combinations. PPIn signal by recruiting proteins via canonical domains or short polybasic motifs. Although their actions are predominantly documented on cytoplasmic membranes, six of the seven PPIn are present within the nucleus together with the PPIn kinases, phosphatases and phospholipases that regulate their turnover. Importantly, the contribution of nuclear PPIn in the regulation of nuclear processes has led to an increased recognition of their importance compared to their more accepted cytoplasmic roles. This review summarises our knowledge on the identification and functional characterisation of nuclear PPIn-effector proteins as well as their mode of interactions, which tend to favour polybasic motifs.
Collapse
|
10
|
Kim SY, Arai Y, Tani T, Takatsuka H, Saito Y, Kawashima T, Kawakami S, Miyawaki A, Nagai T. Simultaneous imaging of multiple cellular events using high-accuracy fluorescence polarization microscopy. Microscopy (Oxf) 2018; 66:110-119. [PMID: 28043995 DOI: 10.1093/jmicro/dfw110] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/30/2016] [Accepted: 12/16/2016] [Indexed: 11/13/2022] Open
Abstract
Förster resonance energy transfer (FRET) has been widely used to design indicators for biomolecules. Conventional FRET-based indicators enable quantitative measurements of analyzes by calculating the ratio between donor and acceptor fluorophores. However, such 'hetero-FRET'-based indicators, which use multiple differently colored fluorophores, restrict the simultaneous use of other colors of fluorescent molecules. To overcome this problem, we developed a 'homo-FRET'-based Ca2+ indicator, W-Cameleon, composed of two identical yellow fluorescent proteins. The binding of Ca2+ to the indicator induces a change in FRET efficiency, which in turn transforms into changes in fluorescence anisotropy. Given that the fluorescence polarization is depolarized by light passing through a high numerical aperture lens and reflecting on a dichroic mirror, we also developed a microscopy technique that reliably detects fluorescence anisotropy with high precision. Our design is aided by photonic-crystal technology, to compensate for the fluorescence depolarization. We thereby succeeded in the simultaneous visualization of three individual intracellular events by using three different fluorescent indicators. Our system may contribute to an expansion of the number of events that can be observed, which will enable a more quantitative understanding of biological phenomena.
Collapse
Affiliation(s)
- Sang-Yeob Kim
- Laboratory for Nanosystems Physiology, Research Institute for Electronic Science, Hokkaido University, Kita 20, Nishi 10, Kita-ku, Sapporo, Hokkaido 001-0020, Japan
| | - Yoshiyuki Arai
- The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan
| | - Tomomi Tani
- Laboratory for Nanosystems Physiology, Research Institute for Electronic Science, Hokkaido University, Kita 20, Nishi 10, Kita-ku, Sapporo, Hokkaido 001-0020, Japan
| | - Hirofumi Takatsuka
- Scientific Solutions Product Development Division, Product Development Department, Olympus Corporation, 2951 Ishikawa-cho, Hachijoji-shi, Tokyo 192-8507, Japan
| | - Yoshiharu Saito
- Scientific Solutions Product Development Division, Product Development Department, Olympus Corporation, 2951 Ishikawa-cho, Hachijoji-shi, Tokyo 192-8507, Japan
| | - Takayuki Kawashima
- Photonic Lattice, Inc., ICR 2F, 6-6-3 Minami-Yoshinari, Aoba, Sendai, Miyagi 989-3204, Japan
| | - Shojiro Kawakami
- Photonic Lattice, Inc., ICR 2F, 6-6-3 Minami-Yoshinari, Aoba, Sendai, Miyagi 989-3204, Japan
| | - Atsushi Miyawaki
- Laboratory for Cell Function and Dynamics, Brain Science Institute, RIKEN, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Takeharu Nagai
- The Institute of Scientific and Industrial Research, Osaka University, 8-1 Mihogaoka, Ibaraki, Osaka 567-0047, Japan
| |
Collapse
|
11
|
A PKC-MARCKS-PI3K regulatory module links Ca2+ and PIP3 signals at the leading edge of polarized macrophages. PLoS One 2018; 13:e0196678. [PMID: 29715315 PMCID: PMC5929533 DOI: 10.1371/journal.pone.0196678] [Citation(s) in RCA: 26] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2018] [Accepted: 04/17/2018] [Indexed: 12/21/2022] Open
Abstract
The leukocyte chemosensory pathway detects attractant gradients and directs cell migration to sites of inflammation, infection, tissue damage, and carcinogenesis. Previous studies have revealed that local Ca2+ and PIP3 signals at the leading edge of polarized leukocytes play central roles in positive feedback loop essential to cell polarization and chemotaxis. These prior studies showed that stimulation of the leading edge Ca2+ signal can strongly activate PI3K, thereby triggering a larger PIP3 signal, but did not elucidate the mechanistic link between Ca2+ and PIP3 signaling. A hypothesis explaining this link emerged, postulating that Ca2+-activated PKC displaces the MARCKS protein from plasma membrane PIP2, thereby releasing sequestered PIP2 to serve as the target and substrate lipid of PI3K in PIP3 production. In vitro single molecule studies of the reconstituted pathway on lipid bilayers demonstrated the feasibility of this PKC-MARCKS-PI3K regulatory module linking Ca2+ and PIP3 signals in the reconstituted system. The present study tests the model predictions in live macrophages by quantifying the effects of: (a) two pathway activators—PDGF and ATP that stimulate chemoreceptors and Ca2+ influx, respectively; and (b) three pathway inhibitors—wortmannin, EGTA, and Go6976 that inhibit PI3K, Ca2+ influx, and PKC, respectively; on (c) four leading edge activity sensors—AKT-PH-mRFP, CKAR, MARCKSp-mRFP, and leading edge area that report on PIP3 density, PKC activity, MARCKS membrane binding, and leading edge expansion/contraction, respectively. The results provide additional evidence that PKC and PI3K are both essential elements of the leading edge positive feedback loop, and strongly support the existence of a PKC-MARCKS-PI3K regulatory module linking the leading edge Ca2+ and PIP3 signals. As predicted, activators stimulate leading edge PKC activity, displacement of MARCKS from the leading edge membrane and increased leading edge PIP3 levels, while inhibitors trigger the opposite effects. Comparison of the findings for the ameboid chemotaxis of leukocytes with recently published findings for the mesenchymal chemotaxis of fibroblasts suggests that some features of the emerging leukocyte leading edge core pathway (PLC-DAG-Ca2+-PKC-MARCKS-PIP2-PI3K-PIP3) may well be shared by all chemotaxing eukaryotic cells, while other elements of the leukocyte pathway may be specialized features of these highly optimized, professional gradient-seeking cells. More broadly, the findings suggest a molecular mechanism for the strong links between phospho-MARCKS and many human cancers.
Collapse
|
12
|
Regulation of PI3K by PKC and MARCKS: Single-Molecule Analysis of a Reconstituted Signaling Pathway. Biophys J 2017; 110:1811-1825. [PMID: 27119641 DOI: 10.1016/j.bpj.2016.03.001] [Citation(s) in RCA: 51] [Impact Index Per Article: 7.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Revised: 02/09/2016] [Accepted: 03/07/2016] [Indexed: 12/19/2022] Open
Abstract
In chemotaxing ameboid cells, a complex leading-edge signaling circuit forms on the cytoplasmic leaflet of the plasma membrane and directs both actin and membrane remodeling to propel the leading edge up an attractant gradient. This leading-edge circuit includes a putative amplification module in which Ca(2+)-protein kinase C (Ca(2+)-PKC) is hypothesized to phosphorylate myristoylated alanine-rich C kinase substrate (MARCKS) and release phosphatidylinositol-4,5-bisphosphate (PIP2), thereby stimulating production of the signaling lipid phosphatidylinositol-3,4,5-trisphosphate (PIP3) by the lipid kinase phosphoinositide-3-kinase (PI3K). We investigated this hypothesized Ca(2+)-PKC-MARCKS-PIP2-PI3K-PIP3 amplification module and tested its key predictions using single-molecule fluorescence to measure the surface densities and activities of its protein components. Our findings demonstrate that together Ca(2+)-PKC and the PIP2-binding peptide of MARCKS modulate the level of free PIP2, which serves as both a docking target and substrate lipid for PI3K. In the off state of the amplification module, the MARCKS peptide sequesters PIP2 and thereby inhibits PI3K binding to the membrane. In the on state, Ca(2+)-PKC phosphorylation of the MARCKS peptide reverses the PIP2 sequestration, thereby releasing multiple PIP2 molecules that recruit multiple active PI3K molecules to the membrane surface. These findings 1) show that the Ca(2+)-PKC-MARCKS-PIP2-PI3K-PIP3 system functions as an activation module in vitro, 2) reveal the molecular mechanism of activation, 3) are consistent with available in vivo data, and 4) yield additional predictions that are testable in live cells. More broadly, the Ca(2+)-PKC-stimulated release of free PIP2 may well regulate the membrane association of other PIP2-binding proteins, and the findings illustrate the power of single-molecule analysis to elucidate key dynamic and mechanistic features of multiprotein signaling pathways on membrane surfaces.
Collapse
|
13
|
Resh MD. Fatty acylation of proteins: The long and the short of it. Prog Lipid Res 2016; 63:120-31. [PMID: 27233110 DOI: 10.1016/j.plipres.2016.05.002] [Citation(s) in RCA: 198] [Impact Index Per Article: 24.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2016] [Revised: 05/19/2016] [Accepted: 05/21/2016] [Indexed: 12/22/2022]
Abstract
Long, short and medium chain fatty acids are covalently attached to hundreds of proteins. Each fatty acid confers distinct biochemical properties, enabling fatty acylation to regulate intracellular trafficking, subcellular localization, protein-protein and protein-lipid interactions. Myristate and palmitate represent the most common fatty acid modifying groups. New insights into how fatty acylation reactions are catalyzed, and how fatty acylation regulates protein structure and function continue to emerge. Myristate is typically linked to an N-terminal glycine, but recent studies reveal that lysines can also be myristoylated. Enzymes that remove N-terminal myristoyl-glycine or myristate from lysines have now been identified. DHHC proteins catalyze S-palmitoylation, but the mechanisms that regulate substrate recognition by individual DHHC family members remain to be determined. New studies continue to reveal thioesterases that remove palmitate from S-acylated proteins. Another area of rapid expansion is fatty acylation of the secreted proteins hedgehog, Wnt and Ghrelin, by Hhat, Porcupine and GOAT, respectively. Understanding how these membrane bound O-acyl transferases recognize their protein and fatty acyl CoA substrates is an active area of investigation, and is punctuated by the finding that these enzymes are potential drug targets in human diseases.
Collapse
Affiliation(s)
- Marilyn D Resh
- Cell Biology Program, Memorial Sloan Kettering Cancer Center, 1275 York Avenue, Box 143, New York, NY 10075, United States.
| |
Collapse
|
14
|
Watching Signaling in Action: Single Molecule Studies of a Reaction Circuit Involved in Chemotaxis. Biophys J 2016; 110:1679-1680. [DOI: 10.1016/j.bpj.2016.03.031] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2016] [Accepted: 03/24/2016] [Indexed: 11/22/2022] Open
|
15
|
In Vitro Neutrophil Migration Requires Protein Kinase C-Delta (δ-PKC)-Mediated Myristoylated Alanine-Rich C-Kinase Substrate (MARCKS) Phosphorylation. Inflammation 2016; 38:1126-41. [PMID: 25515270 DOI: 10.1007/s10753-014-0078-9] [Citation(s) in RCA: 26] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Dysregulated release of neutrophil reactive oxygen species and proteolytic enzymes contributes to both acute and chronic inflammatory diseases. Therefore, molecular regulators of these processes are potential targets for new anti-inflammatory therapies. We have shown previously that myristoylated alanine-rich C-kinase substrate (MARCKS), a well-known actin binding protein and protein kinase C (PKC) substrate, is a key regulator of neutrophil functions. In the current study, we investigate the role of PKC-mediated MARCKS phosphorylation in neutrophil migration and adhesion in vitro. We report that treatment of human neutrophils with the δ-PKC inhibitor rottlerin significantly attenuates f-Met-Leu-Phe (fMLF)-induced MARCKS phosphorylation (IC50=5.709 μM), adhesion (IC50=8.4 μM), and migration (IC50=6.7 μM), while α-, β-, and ζ-PKC inhibitors had no significant effect. We conclude that δ-PKC-mediated MARCKS phosphorylation is essential for human neutrophil migration and adhesion in vitro. These results implicate δ-PKC-mediated MARCKS phosphorylation as a key step in the inflammatory response of neutrophils.
Collapse
|
16
|
Kim DI, Suh BC. Differential interaction of β2e with phosphoinositides: A comparative study between β2e and MARCKS. Channels (Austin) 2015; 10:238-46. [PMID: 26650714 DOI: 10.1080/19336950.2015.1124311] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023] Open
Abstract
Voltage-gated calcium (CaV) channels are responsible for Ca(2+) influx in excitable cells. As one of the auxiliary subunits, the CaV β subunit plays a pivotal role in the membrane expression and receptor modulation of CaV channels. In particular, the subcellular localization of the β subunit is critical for determining the biophysical properties of CaV channels. Recently, we showed that the β2e isotype is tethered to the plasma membrane. Such a feature of β2e is due to the reversible electrostatic interaction with anionic membrane phospholipids. Here, we further explored the membrane interaction property of β2e by comparing it with that of myristoylated alanine-rich C kinase substrate (MARCKS). First, the charge neutralization of the inner leaf of the plasma membrane induced the translocation of both β2e and MARCKS to the cytosol, while the transient depletion of poly-phosphoinositides (poly-PIs) by translocatable pseudojanin (PJ) systems induced the cytosolic translocation of β2e but not MARCKS. Second, the activation of protein kinase C (PKC) induced the translocation of MARCKS but not β2e. We also found that after the cytosolic translocation of MARCKS by receptor activation, depletion of poly-PIs slowed the recovery of MARCKS to the plasma membrane. Together, our data demonstrate that both β2e and MARCKS bind to the membrane through electrostatic interaction but with different binding affinity, and thus, they are differentially regulated by enzymatic degradation of membrane PIs.
Collapse
Affiliation(s)
- Dong-Il Kim
- a Department of Brain and Cognitive Sciences, DGIST , Daegu , Korea
| | - Byung-Chang Suh
- a Department of Brain and Cognitive Sciences, DGIST , Daegu , Korea
| |
Collapse
|
17
|
Rohrbach TD, Jarboe JS, Anderson JC, Trummell HQ, Hicks PH, Weaver AN, Yang ES, Oster RA, Deshane JS, Steele C, Siegal GP, Bonner JA, Willey CD. Targeting the effector domain of the myristoylated alanine rich C-kinase substrate enhances lung cancer radiation sensitivity. Int J Oncol 2014; 46:1079-88. [PMID: 25524703 DOI: 10.3892/ijo.2014.2799] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/30/2014] [Accepted: 11/07/2014] [Indexed: 11/06/2022] Open
Abstract
Lung cancer is the leading cause of cancer related deaths. Common molecular drivers of lung cancer are mutations in receptor tyrosine kinases (RTKs) leading to activation of the phosphatidylinositol 3-kinase (PI3K)/Akt pro-growth, pro-survival signaling pathways. Myristoylated alanine rich C-kinase substrate (MARCKS) is a protein that has the ability to mitigate this signaling cascade by sequestering the target of PI3K, phosphatidylinositol (4,5)-bisphosphate (PIP2). As such, MARCKS has been implicated as a tumor suppressor, though there is some evidence that MARCKS may be tumor promoting in certain cancer types. Since the MARCKS function depends on its phosphorylation status, which impacts its subcellular location, MARCKS role in cancer may depend highly on the signaling context. Currently, the importance of MARCKS in lung cancer biology is limited. Thus, we investigated MARCKS in both clinical specimens and cell culture models. Immunohistochemistry scoring of MARCKS protein expression in a diverse lung tumor tissue array revealed that the majority of squamous cell carcinomas stained positive for MARCKS while other histologies, such as adenocarcinomas, had lower levels. To study the importance of MARCKS in lung cancer biology, we used inducible overexpression of wild-type (WT) and non-phosphorylatable (NP)-MARCKS in A549 lung cancer cells that had a low level of endogenous MARCKS. We found that NP-MARCKS expression, but not WT-MARCKS, enhanced the radiosensitivity of A549 cells in part by inhibiting DNA repair as evidenced by prolonged radiation-induced DNA double strand breaks. We confirmed the importance of MARCKS phosphorylation status by treating several lung cancer cell lines with a peptide mimetic of the phosphorylation domain, the effector domain (ED), which effectively attenuated cell growth as measured by cell index. Thus, the MARCKS ED appears to be an important target for lung cancer therapeutic development.
Collapse
Affiliation(s)
- Timothy D Rohrbach
- Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - John S Jarboe
- Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Joshua C Anderson
- Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Hoa Q Trummell
- Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Patricia H Hicks
- Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Alice N Weaver
- Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Eddy S Yang
- Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Robert A Oster
- Division of Preventive Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Jessy S Deshane
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Chad Steele
- Division of Pulmonary, Allergy and Critical Care Medicine, Department of Medicine, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Gene P Siegal
- Department of Pathology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - James A Bonner
- Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, AL, USA
| | - Christopher D Willey
- Department of Radiation Oncology, The University of Alabama at Birmingham, Birmingham, AL, USA
| |
Collapse
|
18
|
Lippoldt J, Händel C, Dietrich U, Käs J. Dynamic membrane structure induces temporal pattern formation. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2014; 1838:2380-90. [DOI: 10.1016/j.bbamem.2014.05.018] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/27/2014] [Revised: 05/05/2014] [Accepted: 05/16/2014] [Indexed: 10/25/2022]
|
19
|
Graybill C, Prehoda KE. Ordered multisite phosphorylation of lethal giant larvae by atypical protein kinase C. Biochemistry 2014; 53:4931-7. [PMID: 25000553 PMCID: PMC4372073 DOI: 10.1021/bi500748w] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
In Par complex-mediated cell polarity, phosphorylation by atypical protein kinase C (aPKC) is coupled to substrate cortical displacement. Polarized substrates often contain multiple phosphorylation sites, but the role of multisite phosphorylation in Par-mediated polarity remains unclear. Here, we have dissected the role of the three aPKC phosphorylation sites within the tumor suppressor Lethal giant larvae. Using a cultured Drosophila S2 cell cortical displacement assay, we observed that phosphorylation at any one site causes only partial displacement. Complete displacement requires that all three sites be modified. We undertook a kinetic analysis to determine if aPKC phosphorylates each site equivalently. As the sites are closely spaced, we observed not only differences in the rate of phosphorylation but also interaction between the sites. A complete description of the rates reveals a preferential order of phosphorylation. Our results provide new insights into how multiple phosphorylations and phosphorylation rates could regulate localization behaviors of fate determinants at the cortex.
Collapse
Affiliation(s)
- Chiharu Graybill
- The Institute of Molecular Biology and Department of Chemistry and Biochemistry, University of Oregon , Eugene, Oregon 97403, United States
| | | |
Collapse
|
20
|
Tinoco LW, Fraga JL, Anobom CD, Zolessi FR, Obal G, Toledo A, Pritsch O, Arruti C. Structural characterization of a neuroblast-specific phosphorylated region of MARCKS. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2014; 1844:837-49. [PMID: 24590112 DOI: 10.1016/j.bbapap.2014.02.016] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/18/2013] [Revised: 02/07/2014] [Accepted: 02/20/2014] [Indexed: 11/19/2022]
Abstract
MARCKS (Myristoylated Alanine-Rich C Kinase substrate) is a natively unfolded protein that interacts with actin, Ca(2+)-Calmodulin, and some plasma membrane lipids. Such interactions occur at a highly conserved region that is specifically phosphorylated by PKC: the Effector Domain. There are two other conserved domains, MH1 (including a myristoylation site) and MH2, also located in the amino terminal region and whose structure and putative protein binding capabilities are currently unknown. MH2 sequence contains a serine that we described as being phosphorylated only in differentiating neurons (S25 in chick). Here, Circular Dichroism (CD) and Nuclear Magnetic Resonance (NMR) spectroscopy were used to characterize the phosphorylated and unphosphorylated forms of a peptide with the MARCKS sequence surrounding S25. The peptide phosphorylated at this residue is recognized by monoclonal antibody 3C3 (mAb 3C3). CD and NMR data indicated that S25 phosphorylation does not cause extensive modifications in the peptide structure. However, the sharper lines, the absence of multiple spin systems and relaxation dispersion data observed for the phosphorylated peptide suggested a more ordered structure. Surface Plasmon Resonance was employed to compare the binding properties of mAb 3C3 to MARCKS protein and peptide. SPR showed that mAb 3C3 binds to the whole protein and the peptide with a similar affinity, albeit different kinetics. The slightly ordered structure of the phosphorylated peptide might be at the origin of its ability to interact with mAb 3C3 antibody, but this binding did not noticeably modify the peptide structure.
Collapse
Affiliation(s)
- Luzineide W Tinoco
- Instituto de Pesquisas de Produtos Naturais, Universidade Federal do Rio de Janeiro, Cidade Universitária, CCS, Bloco H, Rio de Janeiro 21941-902, RJ, Brazil.
| | - Jully L Fraga
- Instituto de Pesquisas de Produtos Naturais, Universidade Federal do Rio de Janeiro, Cidade Universitária, CCS, Bloco H, Rio de Janeiro 21941-902, RJ, Brazil.
| | - Cristiane D Anobom
- Departamento de Bioquímica, Instituto de Química, Universidade Federal do Rio de Janeiro, Cidade Universitária, CT, Bloco A, Rio de Janeiro 21941-909, RJ, Brazil.
| | - Flavio R Zolessi
- Laboratorio de Cultivo de Tejidos, Sección Biología Celular, DBCM, Facultad de Ciencias, Universidad de la República, Iguá 4225, 11400 Montevideo, Uruguay.
| | - Gonzalo Obal
- Unidad de Biofísica de Proteínas, Institut Pasteur de Montevideo, Mataojo 2020, 11400 Montevideo, Uruguay.
| | - Andrea Toledo
- Laboratorio de Cultivo de Tejidos, Sección Biología Celular, DBCM, Facultad de Ciencias, Universidad de la República, Iguá 4225, 11400 Montevideo, Uruguay.
| | - Otto Pritsch
- Unidad de Biofísica de Proteínas, Institut Pasteur de Montevideo, Mataojo 2020, 11400 Montevideo, Uruguay.
| | - Cristina Arruti
- Laboratorio de Cultivo de Tejidos, Sección Biología Celular, DBCM, Facultad de Ciencias, Universidad de la República, Iguá 4225, 11400 Montevideo, Uruguay.
| |
Collapse
|
21
|
Alonso S, Bär M. Modeling domain formation of MARCKS and protein kinase C at cellular membranes. ACTA ACUST UNITED AC 2014. [DOI: 10.1140/epjnbp14] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
|
22
|
Shimada-Shimizu N, Hisamitsu T, Nakamura TY, Hirayama N, Wakabayashi S. Na+/H+ exchanger 1 is regulated via its lipid-interacting domain, which functions as a molecular switch: a pharmacological approach using indolocarbazole compounds. Mol Pharmacol 2013; 85:18-28. [PMID: 24136992 DOI: 10.1124/mol.113.089268] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
The plasma membrane Na(+)/H(+) exchanger 1 (NHE1) is rapidly activated in response to various stimuli. The membrane-proximal cytoplasmic region (∼60 residues), termed the lipid-interacting domain (LID), is an important regulatory domain of NHE1. Here, we used a pharmacological approach to further characterize the role of LID in the regulation of NHE1. Pharmacological analysis using staurosporine-like indolocarbazole and bisindolylmaleimide compounds suggested that the phorbol ester- and receptor agonist-induced activation of NHE1 occurs through a protein kinase C-independent mechanism. In particular, only indolocarbazole compounds that inhibited NHE1 activation were able to interact with the LID, suggesting that the inhibition of NHE1 activation is achieved through the direct action of these compounds on the LID. Furthermore, in addition to phorbol esters and a receptor agonist, okadaic acid and hyperosmotic stress, which are known to activate NHE1 through unknown mechanisms, were found to promote membrane association of the LID concomitant with NHE1 activation; these effects were inhibited by staurosporine, as well as by a mutation in the LID. Binding experiments using the fluorescent ATP analog trinitrophenyl ATP revealed that ATP and the NHE1 activator phosphatidylinositol 4,5-bisphosphate bind competitively to the LID. These findings suggest that modulation of NHE1 activity by various activators and inhibitors occurs through the direct binding of these molecules to the LID, which alters the association of the LID with the plasma membrane.
Collapse
Affiliation(s)
- Naoko Shimada-Shimizu
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Osaka, Japan (N.S.-S., T.H., T.Y.N., S.W.); and Basic Medical Science & Molecular Medicine, Tokai University School of Medicine, Kanagawa, Japan (N.H.)
| | | | | | | | | |
Collapse
|
23
|
Ott LE, Sung EJ, Melvin AT, Sheats MK, Haugh JM, Adler KB, Jones SL. Fibroblast Migration Is Regulated by Myristoylated Alanine-Rich C-Kinase Substrate (MARCKS) Protein. PLoS One 2013; 8:e66512. [PMID: 23840497 PMCID: PMC3686679 DOI: 10.1371/journal.pone.0066512] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2012] [Accepted: 05/10/2013] [Indexed: 01/10/2023] Open
Abstract
Myristoylated alanine-rich C-kinase substrate (MARCKS) is a ubiquitously expressed substrate of protein kinase C (PKC) that is involved in reorganization of the actin cytoskeleton. We hypothesized that MARCKS is involved in regulation of fibroblast migration and addressed this hypothesis by utilizing a unique reagent developed in this laboratory, the MANS peptide. The MANS peptide is a myristoylated cell permeable peptide corresponding to the first 24-amino acids of MARCKS that inhibits MARCKS function. Treatment of NIH-3T3 fibroblasts with the MANS peptide attenuated cell migration in scratch wounding assays, while a myristoylated, missense control peptide (RNS) had no effect. Neither MANS nor RNS peptide treatment altered NIH-3T3 cell proliferation within the parameters of the scratch assay. MANS peptide treatment also resulted in inhibited NIH-3T3 chemotaxis towards the chemoattractant platelet-derived growth factor-BB (PDGF-BB), with no effect observed with RNS treatment. Live cell imaging of PDGF-BB induced chemotaxis demonstrated that MANS peptide treatment resulted in weak chemotactic fidelity compared to RNS treated cells. MANS and RNS peptides did not affect PDGF-BB induced phosphorylation of MARCKS or phosphoinositide 3-kinase (PI3K) signaling, as measured by Akt phosphorylation. Further, no difference in cell migration was observed in NIH-3T3 fibroblasts that were transfected with MARCKS siRNAs with or without MANS peptide treatment. Genetic structure-function analysis revealed that MANS peptide-mediated attenuation of NIH-3T3 cell migration does not require the presence of the myristic acid moiety on the amino-terminus. Expression of either MANS or unmyristoylated MANS (UMANS) C-terminal EGFP fusion proteins resulted in similar levels of attenuated cell migration as observed with MANS peptide treatment. These data demonstrate that MARCKS regulates cell migration and suggests that MARCKS-mediated regulation of fibroblast migration involves the MARCKS amino-terminus. Further, this data demonstrates that MANS peptide treatment inhibits MARCKS function during fibroblast migration and that MANS mediated inhibition occurs independent of myristoylation.
Collapse
Affiliation(s)
- Laura E. Ott
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
- Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Eui Jae Sung
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
- Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Adam T. Melvin
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Mary K. Sheats
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
- Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Jason M. Haugh
- Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina, United States of America
- Department of Chemical and Biomolecular Engineering, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Kenneth B. Adler
- Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina, United States of America
- Department of Molecular Biomedical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
| | - Samuel L. Jones
- Department of Clinical Sciences, College of Veterinary Medicine, North Carolina State University, Raleigh, North Carolina, United States of America
- Center for Comparative Medicine and Translational Research, North Carolina State University, Raleigh, North Carolina, United States of America
- * E-mail:
| |
Collapse
|
24
|
Wuttke A, Idevall-Hagren O, Tengholm A. P2Y₁ receptor-dependent diacylglycerol signaling microdomains in β cells promote insulin secretion. FASEB J 2013; 27:1610-20. [PMID: 23299857 DOI: 10.1096/fj.12-221499] [Citation(s) in RCA: 41] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Diacylglycerol (DAG) controls numerous cell functions by regulating the localization of C1-domain-containing proteins, including protein kinase C (PKC), but little is known about the spatiotemporal dynamics of the lipid. Here, we explored plasma membrane DAG dynamics in pancreatic β cells and determined whether DAG signaling is involved in secretagogue-induced pulsatile release of insulin. Single MIN6 cells, primary mouse β cells, and human β cells within intact islets were transfected with translocation biosensors for DAG, PKC activity, or insulin secretion and imaged with total internal reflection fluorescence microscopy. Muscarinic receptor stimulation triggered stable, homogenous DAG elevations, whereas glucose induced short-lived (7.1 ± 0.4 s) but high-amplitude elevations (up to 109 ± 10% fluorescence increase) in spatially confined membrane regions. The spiking was mimicked by membrane depolarization and suppressed after inhibition of exocytosis or of purinergic P2Y₁, but not P2X receptors, reflecting involvement of autocrine purinoceptor activation after exocytotic release of ATP. Each DAG spike caused local PKC activation with resulting dissociation of its substrate protein MARCKS from the plasma membrane. Inhibition of spiking reduced glucose-induced pulsatile insulin secretion. Thus, stimulus-specific DAG signaling patterns appear in the plasma membrane, including distinct microdomains, which have implications for the kinetic control of exocytosis and other membrane-associated processes.
Collapse
Affiliation(s)
- Anne Wuttke
- Department of Medical Cell Biology, Uppsala University, Biomedical Centre, Uppsala, Sweden
| | | | | |
Collapse
|
25
|
Dimatelis JJ, Russell VA, Stein DJ, Daniels WM. Effects of maternal separation and methamphetamine exposure on protein expression in the nucleus accumbens shell and core. Metab Brain Dis 2012; 27:363-75. [PMID: 22451087 DOI: 10.1007/s11011-012-9295-9] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/29/2011] [Accepted: 03/15/2012] [Indexed: 12/14/2022]
Abstract
Early life adversity has been suggested to predispose an individual to later drug abuse. The core and shell sub-regions of the nucleus accumbens are differentially affected by both stressors and methamphetamine. This study aimed to characterize and quantify methamphetamine-induced protein expression in the shell and core of the nucleus accumbens in animals exposed to maternal separation during early development. Isobaric tagging (iTRAQ) which enables simultaneous identification and quantification of peptides with tandem mass spectrometry (MS/MS) was used. We found that maternal separation altered more proteins involved in structure and redox regulation in the shell than in the core of the nucleus accumbens, and that maternal separation and methamphetamine had differential effects on signaling proteins in the shell and core. Compared to maternal separation or methamphetamine alone, the maternal separation/methamphetamine combination altered more proteins involved in energy metabolism, redox regulatory processes and neurotrophic proteins. Methamphetamine treatment of rats subjected to maternal separation caused a reduction of cytoskeletal proteins in the shell and altered cytoskeletal, signaling, energy metabolism and redox proteins in the core. Comparison of maternal separation/methamphetamine to methamphetamine alone resulted in decreased cytoskeletal proteins in both the shell and core and increased neurotrophic proteins in the core. This study confirms that both early life stress and methamphetamine differentially affect the shell and core of the nucleus accumbens and demonstrates that the combination of early life adversity and later methamphetamine use results in more proteins being affected in the nucleus accumbens than either treatment alone.
Collapse
Affiliation(s)
- J J Dimatelis
- Department of Human Biology, Faculty of Health Sciences, University of Cape Town, Observatory, 7925 Cape Town, South Africa.
| | | | | | | |
Collapse
|
26
|
Paramanik V, Thakur MK. Estrogen receptor β and its domains interact with casein kinase 2, phosphokinase C, and N-myristoylation sites of mitochondrial and nuclear proteins in mouse brain. J Biol Chem 2012; 287:22305-16. [PMID: 22566700 DOI: 10.1074/jbc.m112.351262] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The localization of estrogen receptor (ER)β in mitochondria suggests ERβ-dependent regulation of genes, which is poorly understood. Here, we analyzed the ERβ interacting mitochondrial as well as nuclear proteins in mouse brain using pull-down assay and matrix-assisted laser desorption ionization mass spectroscopy (MALDI-MS). In the case of mitochondria, ERβ interacted with six proteins of 35-152 kDa, its transactivation domain (TAD) interacted with four proteins of 37-172 kDa, and ligand binding domain (LBD) interacted with six proteins of 37-161 kDa. On the other hand, in nuclei, ERβ interacted with seven proteins of 30-203 kDa, TAD with ten proteins of 31-160 kDa, and LBD with fourteen proteins of 42-179 kDa. For further identification, these proteins were cleaved by trypsin into peptides and analyzed by MALDI-MS using mascot search engine, immunoprecipitation, immunoblotting, and far-Western blotting. To find the consensus binding motifs in interacting proteins, their unique tryptic peptides were analyzed by the motif scan software. All the interacting proteins were found to contain casein kinase (CK) 2, phosphokinase (PK)C phosphorylation, and N-myristoylation sites. These were further confirmed by peptide pull-down assays using specific mutations in the interacting sites. Thus, the present findings provide evidence for the interaction of ERβ with specific mitochondrial and nuclear proteins through consensus CK2, PKC phosphorylation, and N-myristoylation sites, and may represent an essential step toward designing selective ER modulators for regulating estrogen-mediated signaling.
Collapse
Affiliation(s)
- Vijay Paramanik
- Biochemistry and Molecular Biology Laboratory, Centre of Advanced Study, Department of Zoology, Banaras Hindu University, Varanasi-221 005, India
| | | |
Collapse
|
27
|
A Role for the PKC Signaling System in the Pathophysiology and Treatment of Mood Disorders: Involvement of a Functional Imbalance? Mol Neurobiol 2011; 44:407-19. [DOI: 10.1007/s12035-011-8210-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2011] [Accepted: 09/20/2011] [Indexed: 12/15/2022]
|
28
|
Alonso S, Dietrich U, Händel C, Käs JA, Bär M. Oscillations in the lateral pressure of lipid monolayers induced by nonlinear chemical dynamics of the second messengers MARCKS and protein kinase C. Biophys J 2011; 100:939-47. [PMID: 21320438 DOI: 10.1016/j.bpj.2010.12.3702] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2010] [Revised: 11/26/2010] [Accepted: 12/02/2010] [Indexed: 11/29/2022] Open
Abstract
The binding of the MARCKS peptide to the lipid monolayer containing PIP(2) increases the lateral pressure of the monolayer. The unbinding dynamics modulated by protein kinase C leads to oscillations in lateral pressure of lipid monolayers. These periodic dynamics can be attributed to changes in the crystalline lipid domain size. We have developed a mathematical model to explain these observations based on the changes in the physical structure of the monolayer by the translocation of MARCKS peptide. The model indicates that changes in lipid domain size drives these oscillations. The model is extended to an open system that sustains chemical oscillations.
Collapse
Affiliation(s)
- Sergio Alonso
- Physikalisch-Technische Bundesanstalt, Berlin, Germany.
| | | | | | | | | |
Collapse
|
29
|
Dietrich U, Krüger P, Käs JA. Structural investigation on the adsorption of the MARCKS peptide on anionic lipid monolayers - effects beyond electrostatic. Chem Phys Lipids 2011; 164:266-75. [PMID: 21376024 DOI: 10.1016/j.chemphyslip.2011.02.004] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 02/16/2011] [Accepted: 02/17/2011] [Indexed: 11/25/2022]
Abstract
The presence of charged lipids in the cell membrane constitutes the background for the interaction with numerous membrane proteins. As a result, the valence of the lipids plays an important role concerning their lateral organization in the membrane and therefore the very manner of this interaction. This present study examines this aspect, particularly regarding to the interaction of the anionic lipid DPPS with the highly basic charged effector domain of the MARCKS protein, examined in monolayer model systems. Film balance, fluorescence microscopy and X-ray reflection/diffraction measurements were used to study the behavior of DPPS in a mixture with DPPC for its dependance on the presence of MARCKS (151-175). In the mixed monolayer, both lipids are completely miscible therefore DPPS is incorporated in the ordered crystalline DPPC domains as well. The interaction of MARCKS peptide with the mixed monolayer leads to the formation of lipid/peptide clusters causing an elongation of the serine group of the DPPS up to 7Å in direction to surface normal into the subphase. The large cationic charge of the peptide pulls out the serine group of the interface which simultaneously causes an elongation of the phosphodiester group of the lipid fraction too. The obtained results were used to compare the interaction of MARCKS peptide with the polyvalent PIP(2) in mixed monolayers. On this way we surprisingly find out, that the relative small charge difference of the anionic lipids causes a significant different interaction with MARCKS (151-175). The lateral arrangement of the anionic lipids depends on their charge values and determines the diffusion of the electrostatic binding clusters within the membrane.
Collapse
Affiliation(s)
- Undine Dietrich
- Division of Soft Matter Physics, Leipzig University, Linnstrasse, Germany.
| | | | | |
Collapse
|
30
|
Green TD, Crews AL, Park J, Fang S, Adler KB. Regulation of mucin secretion and inflammation in asthma: a role for MARCKS protein? Biochim Biophys Acta Gen Subj 2011; 1810:1110-3. [PMID: 21281703 DOI: 10.1016/j.bbagen.2011.01.009] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2010] [Revised: 01/14/2011] [Accepted: 01/21/2011] [Indexed: 10/18/2022]
Abstract
BACKGROUND A major characteristic of asthmatic airways is an increase in mucin (the glycoprotein component of mucus) producing and secreting cells, which leads to increased mucin release that further clogs constricted airways and contributes markedly to airway obstruction and, in the most severe cases, to status asthmaticus. Asthmatic airways show both a hyperplasia and metaplasia of goblet cells, mucin-producing cells in the epithelium; hyperplasia refers to enhanced numbers of goblet cells in larger airways, while metaplasia refers to the appearance of these cells in smaller airways where they normally are not seen. With the number of mucin-producing and secreting cells increased, there is a coincident hypersecretion of mucin which characterizes asthma. On a cellular level, a major regulator of airway mucin secretion in both in vitro and in vivo studies has been shown to be MARCKS (myristoylated alanine-rich C kinase substrate) protein, a ubiquitous substrate of protein kinase C (PKC). GENERAL SIGNIFICANCE In this review, properties of MARCKS and how the protein may regulate mucin secretion at a cellular level will be discussed. In addition, the roles of MARCKS in airway inflammation related to both influx of inflammatory cells into the lung and release of granules containing inflammatory mediators by these cells will be explored. This article is part of a Special Issue entitled: Biochemistry of Asthma.
Collapse
Affiliation(s)
- Teresa D Green
- Deparment of Molecualr Biomedical Sciences, North Carolina State University CVM, Raleigh, NC 27606, USA
| | | | | | | | | |
Collapse
|
31
|
Alonso S, Bär M. Phase separation and bistability in a three-dimensional model for protein domain formation at biomembranes. Phys Biol 2010; 7:046012. [PMID: 21149929 DOI: 10.1088/1478-3975/7/4/046012] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Proteins in living cells interact with membranes. They may bind to or unbind from the membrane to the cytosol depending on the lipid composition of the membrane and their interaction with cytosolic enzymes. Moreover, proteins can accumulate at the membrane and assemble in spatial domains. Here, a simple model of protein cycling at biomembranes is studied, when the total number of proteins is conserved. Specifically, we consider the spatio-temporal dynamics of MARCKS proteins and their interactions with enzymes facilitating translocation from and rebinding to the membrane. The model exhibits two qualitatively different mechanisms of protein domain formation: phase separation related to a long-wave instability of a membrane state with homogeneous protein coverage and stable coexistence of two states with different homogeneous protein coverage in bistable media. We evaluate the impact of the cytosolic volume on the occurrence of protein pattern formation by simulations in a three-dimensional model. We show that the explicit treatment of the volume in the model leads to an effective rescaling of the reaction rates. For a simplified model of protein cycling, we can derive analytical expressions for the rescaling coefficients and verify them by direct simulations with the complete three-dimensional model.
Collapse
Affiliation(s)
- Sergio Alonso
- Physikalisch-Technische Bundesanstalt, Abbestrasse 2-12, 10587 Berlin, Germany.
| | | |
Collapse
|
32
|
Si FC. Effects of Qigesan, Shashenmaidongtang, Tongyoutang and Buqiyunpitang on hEGF-stimulated proliferation of human esophageal carcinoma EC9706 cells. Shijie Huaren Xiaohua Zazhi 2010; 18:2956-2965. [DOI: 10.11569/wcjd.v18.i28.2956] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To investigate the effects of Qigesan, Shashenmaidongtang, Tongyoutang and Buqiyunpitang on the proliferation of human esophageal carcinoma EC9706 cells stimulated with human epidermal growth factor (hEGF), and to explore potential mechanisms involved.
METHODS: After cultured EC9706 cells were stimulated with hEGF and then treated with Qigesan, Shashenmaidongtang, Tongyoutang and Buqiyunpitang, respectively, cell morphological changes were observed under an inverted microscope, cell proliferation was determined by MTT assay, cell cycle progression was measured by flow cytometry, and protein expression and tyrosine phosphorylation in PLC-γ1 and PI3K-mediated signaling pathways were determined by Western blot.
RESULTS: The half-maximum inhibitory concentrations (IC50) of Qigesan, Shashenmaidongtang and Tongyoutang were 849, 1 004 and 1 615 mg/L, respectively. These drugs at a concentration of IC50 inhibited hEGF-stimulated cell proliferation and prevented cell cycle progression into S phase. Buqiyunpitang could only weakly inhibit the proliferation of EC9706 cells. Qigesan, Shashenmaidongtang and Tongyoutang inhibited the protein expression and tyrosine phosphorylation of EGFR and PLC-γ1 as well as the protein expression of PKCα, MARCKS, PI3K, AKT-1 and NF-κB p50. Tongyoutang had the strongest inhibitory effects on the PLC-γ1 signaling pathway, followed by Qigesan and Shashenmaidongtang. In contrast, the order of the inhibitory potency for the PI3K signaling pathway was Tongyoutang, Shashenmaidongtang and Qigesan.
CONCLUSION: Qigesan, Shashenmaidongtang and Tongyoutang can, to varying degrees, inhibit hEGF-stimulated cell proliferation by suppressing PLC-γ1 and PI3K-mediated signaling pathways.
Collapse
|
33
|
Wakabayashi S, Nakamura TY, Kobayashi S, Hisamitsu T. Novel phorbol ester-binding motif mediates hormonal activation of Na+/H+ exchanger. J Biol Chem 2010; 285:26652-61. [PMID: 20551318 DOI: 10.1074/jbc.m110.130120] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Protein kinase C (PKC) is considered crucial for hormonal Na(+)/H(+) exchanger (NHE1) activation because phorbol esters (PEs) strongly activate NHE1. However, here we report that rather than PKC, direct binding of PEs/diacylglycerol to the NHE1 lipid-interacting domain (LID) and the subsequent tighter association of LID with the plasma membrane mainly underlies NHE1 activation. We show that (i) PEs directly interact with the LID of NHE1 in vitro, (ii) like PKC, green fluorescent protein (GFP)-labeled LID translocates to the plasma membrane in response to PEs and receptor agonists, (iii) LID mutations markedly inhibit these interactions and PE/receptor agonist-induced NHE1 activation, and (iv) PKC inhibitors ineffectively block NHE1 activation, except staurosporin, which itself inhibits NHE1 via LID. Thus, we propose a PKC-independent mechanism of NHE1 regulation via a PE-binding motif previously unrecognized.
Collapse
Affiliation(s)
- Shigeo Wakabayashi
- Department of Molecular Physiology, National Cerebral and Cardiovascular Center Research Institute, Suita, Osaka, Japan.
| | | | | | | |
Collapse
|
34
|
Homchaudhuri L, Polverini E, Gao W, Harauz G, Boggs JM. Influence of membrane surface charge and post-translational modifications to myelin basic protein on its ability to tether the Fyn-SH3 domain to a membrane in vitro. Biochemistry 2010; 48:2385-93. [PMID: 19178193 DOI: 10.1021/bi8022587] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
Myelin basic protein (MBP) is a highly post-translationally modified, multifunctional structural component of central nervous system myelin, adhering to phospholipid membranes and assembling cytoskeletal proteins, and has previously been shown to bind SH3 domains in vitro and tether them to a membrane surface [Polverini, E., et al. (2008) Biochemistry 47, 267-282]. Since molecular modeling shows that the Fyn-SH3 domain has a negative surface charge density even after binding the MBP ligand, we have investigated the influence of negative membrane surface charge and the effects of post-translational modifications to MBP on the interaction of the Fyn-SH3 domain with membrane-associated MBP. Using a sedimentation assay with multilamellar vesicles consisting of neutral phosphatidylcholine (PC) and negatively charged phosphatidylinositol (PI), we demonstrate that increasing the negative surface charge of the membrane by increasing the proportion of PI reduces the amount of Fyn-SH3 domain that binds to membrane-associated MBP, due to electrostatic repulsion. When one of the phosphoinositides, PI(4)P or PI(4,5)P(2) was substituted for PI in equal proportion, none of the Fyn-SH3 domain bound to MBP under the conditions that were used. Post-translational modifications of MBP which reduced its net positive charge, i.e., phosphorylation or arginine deimination, increased the degree of repulsion of Fyn-SH3 from the membrane surface, an effect further modulated by the lipid charge. This study suggests that changes in membrane negative surface charge due to protein or lipid modifications, which could occur during cell signaling, can regulate the binding of the Fyn-SH3 domain to membrane-associated MBP and thus could regulate the activity of Fyn at the oligodendrocyte membrane surface.
Collapse
Affiliation(s)
- Lopamudra Homchaudhuri
- Department of Molecular Structure and Function, Research Institute, Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| | | | | | | | | |
Collapse
|
35
|
Adachi E, Kazoe Y, Sato Y, Suzuki Y, Urano T, Ueyama T, Saito N, Nikolaev VO, Lohse MJ, Tominaga M, Mogami H. A technique for monitoring multiple signals with a combination of prism-based total internal reflection fluorescence microscopy and epifluorescence microscopy. Pflugers Arch 2009; 459:227-34. [PMID: 19680684 DOI: 10.1007/s00424-009-0705-8] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/13/2009] [Revised: 07/08/2009] [Accepted: 07/21/2009] [Indexed: 10/20/2022]
Abstract
Physiological phenomena are regulated by multiple signal pathways upon receptor stimulation. Here, we have introduced a new technique with a combination of prism-based total internal reflection fluorescence microscopy (PBTIRFM) and epifluorescence microscopy (EPI) to simultaneously monitor multiple signal pathways. This instrumentation allows us to visualize three signal pathways, Ca2+, cyclic adenosine monophosphate (cAMP)/protein kinase A (PKA), and diacylglycerol (DAG)/protein kinase C (PKC) signals in living cells. Three fluorescent indicators were employed for this purpose: (1) Fura-2 AM as a calcium sensor; (2) Epac1-camp, a cyan fluorescent protein-yellow fluorescent protein fluorescence resonance energy transfer-based cAMP indicator, as a cAMP sensor; and (3) C1-tagged monomeric red fluorescent protein, a tandem DAG-binding domain of PKC gamma, as a DAG sensor or myristoylated alanine-rich C kinase substrate-tagged DsRed for the PKC activation pathway. The DAG signal was monitored by PBTIRFM, whereas the Ca2+ and cAMP signals were monitored by EPI. Adenosine trisphosphate resulted in generation of all three second messengers in triple probe-loaded Cos-7 cells. The spectral overlap between these signal probes was evaluated by means of linear unmixing. Forskolin also evoked Ca2+, cAMP/PKA, and DAG/PKC signals whereas acetylcholine activated Ca2+ and DAG/PKC signals as well as inhibiting cAMP generation in triple probe-loaded insulin-secreting cells. Thus, the optical observation system combining PBTIRFM and EPI offers a great advance in analyzing interplay of multiple signaling pathways, such as these second messengers, upon G-protein-coupled receptor stimulation in living cells.
Collapse
Affiliation(s)
- Eisuke Adachi
- Department of Physiology, Hamamatsu University School of Medicine, Handa-yama 1-20-1, Higashi-ku, Hamamatsu, Shizuoka, 431-3192, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
36
|
Pores-Fernando AT, Ranaghan MYD, Zweifach A. No specific subcellular localization of protein kinase C is required for cytotoxic T cell granule exocytosis. J Biol Chem 2009; 284:25107-15. [PMID: 19592494 DOI: 10.1074/jbc.m109.011866] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Cytotoxic T cells kill virus-infected cells and tumor cells by releasing lytic granules that contain cell-killing contents. Exocytosis requires calcium influx and protein kinase C (PKC) activation. Here, we extend our previous finding regarding the lack of isoform specificity of PKCs in the granule release step, showing that mutant constitutively active PKCdelta can substitute for phorbol esters and support exocytosis. PKCdelta, a novel PKC isoform, was recently shown to play a role in lytic granule reorientation. Surprisingly, however, our results suggested that mutant PKCdelta did not localize to the plasma membrane (PM). To test directly whether PKC has to be in the PM to drive exocytosis, we generated mutants of various PKC isoforms that were tethered either to the outer mitochondrial membrane or to the PM. Tethered mutant PKCdeltas were able to promote exocytosis as effectively as the untethered version. The substrates of PKCs involved in lytic granule exocytosis are currently unknown, but subcellular localization is believed to be a critical factor in determining PKC accessibility to substrates. That there is no requirement for specific PKC localization in lytic granule exocytosis may have important implications for the identity of PKC substrates.
Collapse
Affiliation(s)
- Arun T Pores-Fernando
- Department of Molecular and Cell Biology, University of Connecticut, Storrs, Connecticut 06269-3125, USA
| | | | | |
Collapse
|
37
|
Estrada-Bernal A, Gatlin JC, Sunpaweravong S, Pfenninger KH. Dynamic adhesions and MARCKS in melanoma cells. J Cell Sci 2009; 122:2300-10. [PMID: 19509053 DOI: 10.1242/jcs.047860] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Cell motility necessitates the rapid formation and disassembly of cell adhesions. We have studied adhesions in a highly motile melanoma cell line using various biochemical approaches and microscopic techniques to image close adhesions. We report that WM-1617 melanoma cells contain at least two types of close adhesion: classic focal adhesions and more extensive, irregularly shaped adhesions that tend to occur along lamellipodial edges. In contrast to focal adhesions, these latter adhesions are highly dynamic and can be disassembled rapidly via protein kinase C (PKC) activation (e.g. by eicosanoid) and MARCKS phosphorylation. MARCKS overexpression, however, greatly increases the area of close adhesions and renders them largely refractory to PKC stimulation. This indicates that nonphosphorylated MARCKS is an adhesion stabilizer. Unlike focal adhesions, the dynamic adhesions contain alpha3 integrin and MARCKS, but they do not contain the focal adhesion marker vinculin. Overall, these results begin to define the molecular and functional properties of dynamic close adhesions involved in cell motility.
Collapse
Affiliation(s)
- Adriana Estrada-Bernal
- Department of Pediatrics, University of Colorado School of Medicine, University of Colorado Cancer Center, and Colorado Intellectual and Developmental Disabilities Research Center, Aurora, CO 80045, USA
| | | | | | | |
Collapse
|
38
|
Dietrich U, Krüger P, Gutberlet T, Käs JA. Interaction of the MARCKS peptide with PIP2 in phospholipid monolayers. BIOCHIMICA ET BIOPHYSICA ACTA-BIOMEMBRANES 2009; 1788:1474-81. [PMID: 19362071 DOI: 10.1016/j.bbamem.2009.04.001] [Citation(s) in RCA: 30] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/30/2009] [Revised: 03/15/2009] [Accepted: 04/01/2009] [Indexed: 11/17/2022]
Abstract
In this present work we have studied the effect of MARCKS (151-175) peptide on a mixed DPPC/PIP2 monolayer. By means of film balance, fluorescence microscopy, x-ray reflection/diffraction and neutron reflection measurements we detected changes in the lateral organization of the monolayer and changes in the perpendicular orientation of the PIP2 molecules depending on the presence of MARCKS (151-175) peptide in the subphase. In the mixed monolayer, the PIP2 molecules are distributed uniformly in the disordered phase of the monolayer, whereas the PI(4,5) groups elongate up to 10 A below the phosphodiester groups. This elongation forms the precondition for the electrostatic interaction of the MARCKS peptide with the PIP2 molecules. Due to the enrichment of PIP2 in the disordered phase, the interaction with the peptide occurs primarily in this phase, causing the PI(4,5) groups to tilt toward the monolayer interface.
Collapse
Affiliation(s)
- Undine Dietrich
- Division of Soft Matter Physics, Faculty for Physics and Earth Sciences, University of Leipzig, Linnéstr. 5, D-04103 Leipzig, Germany.
| | | | | | | |
Collapse
|
39
|
Sprouty4 negatively regulates protein kinase C activation by inhibiting phosphatidylinositol 4,5-biphosphate hydrolysis. Oncogene 2009; 28:1076-88. [PMID: 19137008 DOI: 10.1038/onc.2008.464] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Sproutys have been shown to negatively regulate growth factor-induced extracellular signal-regulated kinase (ERK) activation, and suggested to be an anti-oncogene. However, molecular mechanism of the suppression has not yet been clarified completely. Sprouty4 inhibits vascular endothelial growth factor (VEGF)-A-induced ERK activation, but not VEGF-C-induced ERK activation. It has been shown that VEGF-A-mediated ERK activation is strongly dependent on protein kinase C (PKC), whereas that by VEGF-C is dependent on Ras. This suggests that Sprouty4 inhibits the PKC pathway more specifically than the Ras pathway. In this study, we confirmed that Sprouty4 suppressed various signals downstream of PKC, such as phosphorylation of MARCKS and protein kinase D (PKD), as well as PKC-dependent nuclear factor (NF)-kappaB activation. Furthermore, Sprouty4 suppressed upstream signals of PKC, such as Ca(2+) mobilization, phosphatidylinositol 4,5-biphosphate (PIP(2)) breakdown and inositol 1,4,5-triphosphate (IP(3)) production in response to VEGF-A. Those effects were dependent on the C-terminal cysteine-rich region, but not on the N-terminal region of Sprouty4, which is critical for the suppression of fibroblast growth factor (FGF)-mediated ERK activation. Sprouty4 overexpression or deletion of the Sprouty4 gene did not affect phospholipase C (PLC) gamma-1 activation, which is an enzyme that catalyzes PIP(2) hydrolysis. Moreover, Sprouty4 inhibited not only VEGF-A-mediated PIP(2) hydrolysis but also inhibited the lysophosphatidic acid (LPA)-induced PIP(2) breakdown that is catalyzed by PLC beta/epsilon activated by G-protein coupled receptor (GPCR). Taken together, Sprouty4 has broader suppression activity for various stimuli than previously thought; it may function as an inhibitor for various types of PLC-dependent signaling as well as for ERK activation.
Collapse
|
40
|
Johnson CM, Rodgers W. Spatial Segregation of Phosphatidylinositol 4,5-Bisphosphate (PIP(2)) Signaling in Immune Cell Functions. IMMUNOLOGY, ENDOCRINE & METABOLIC AGENTS IN MEDICINAL CHEMISTRY 2008; 8:349-357. [PMID: 19956793 PMCID: PMC2771939 DOI: 10.2174/187152208787169233] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Phosphatidylinositol 4,5-bisphosphate (PIP(2)) is a prevalent phosphoinositide in the inner leaflet of the plasma membrane. PIP(2) associates with an ever-growing list of proteins, and participates in a variety of cellular processes. PIP(2) signaling to the actin cytoskeleton transduces specific signals necessary for changes in morphology, motility, endocytosis, exocytosis, phagocytosis, and cell activation. The mechanism(s) by which PIP(2) signaling pathways are specific is a topic of intense investigation. One working model is the compartmentalization of PIP(2)-mediated signaling by concentrating PIP(2) in cholesterol-dependent membrane rafts, therefore providing spatial and temporal regulation. Here we discuss properties of PIP(2) signaling to the actin cytoskeleton in immune cell functioning, the association of PIP(2) cellular pools with membrane rafts, and recent work investigating models for compartmentalization of PIP(2)-mediated signaling in membrane rafts to the actin cytoskeleton.
Collapse
Affiliation(s)
- Corey M. Johnson
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation
| | - William Rodgers
- Cardiovascular Biology Research Program, Oklahoma Medical Research Foundation
- Departments of Microbiology and Immunology, & Pathology, University of Oklahoma Health Sciences Center
| |
Collapse
|
41
|
Musse AA, Gao W, Homchaudhuri L, Boggs JM, Harauz G. Myelin basic protein as a "PI(4,5)P2-modulin": a new biological function for a major central nervous system protein. Biochemistry 2008; 47:10372-82. [PMID: 18767817 DOI: 10.1021/bi801302b] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
The 18.5 kDa isoform of myelin basic protein (MBP) is multifunctional and has previously been shown to have structural and phenomenological similarities with domains of other membrane- and cytoskeleton-associated proteins such as MARCKS (myristoylated alanine-rich C kinase substrate). Here, we have investigated whether 18.5 kDa MBP can sequester phosphatidylinositol-(4,5)-bis-phosphate (PI(4,5)P 2) in membranes, like MARCKS and other "PIPmodulins" do. Using fluorescence-quenching and electron paramagnetic resonance (EPR) spectroscopy, and model membranes containing BODIPY-FL- or proxyl-labeled PI(4,5)P 2, respectively, we have demonstrated that MBP laterally sequesters PI(4,5)P 2. The MBP-PI(4,5)P 2 interactions are electrostatic, partially cholesterol-dependent, and sensitive to phosphorylation, deimination, and Ca (2+)-CaM binding. Confocal microscopy of cultured oligodendrocytes also revealed patched colocalization of MBP and PI(4,5)P 2, indicating the spatial clustering of PI(4,5)P 2 in the plasma membrane. On the basis of these findings as well as the overwhelming convergence of functional properties, modifying enzymes, and interaction partners, we propose that MBP is mechanistically related to GAP-43, MARCKS, and CAP-23. During myelinogenesis, it may mediate calcium and phosphorylation-sensitive plasma membrane availability of PI(4,5)P 2. This regulation of PI(4,5)P 2 availability at the cell cortex may be coupled to the elaboration and outgrowth of the membranous cellular processes by oligodendrocytes.
Collapse
Affiliation(s)
- Abdiwahab A Musse
- Department of Molecular and Cellular Biology and Biophysics Interdepartmental Group, University of Guelph, 50 Stone Road East, Guelph, Ontario, Canada, N1G 2W1
| | | | | | | | | |
Collapse
|
42
|
Pinchuk GV, Lee SR, Nanduri B, Honsinger KL, Stokes JV, Pinchuk LM. Bovine viral diarrhea viruses differentially alter the expression of the protein kinases and related proteins affecting the development of infection and anti-viral mechanisms in bovine monocytes. BIOCHIMICA ET BIOPHYSICA ACTA-PROTEINS AND PROTEOMICS 2008; 1784:1234-47. [DOI: 10.1016/j.bbapap.2008.05.004] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/28/2007] [Revised: 05/05/2008] [Accepted: 05/06/2008] [Indexed: 10/22/2022]
|
43
|
Proteomic modeling for HIV-1 infected microglia-astrocyte crosstalk. PLoS One 2008; 3:e2507. [PMID: 18575609 PMCID: PMC2429966 DOI: 10.1371/journal.pone.0002507] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2008] [Accepted: 05/15/2008] [Indexed: 12/20/2022] Open
Abstract
Background HIV-1-infected and immune competent brain mononuclear phagocytes (MP; macrophages and microglia) secrete cellular and viral toxins that affect neuronal damage during advanced disease. In contrast, astrocytes can affect disease by modulating the nervous system's microenvironment. Interestingly, little is known how astrocytes communicate with MP to influence disease. Methods and Findings MP-astrocyte crosstalk was investigated by a proteomic platform analysis using vesicular stomatitis virus pseudotyped HIV infected murine microglia. The microglial-astrocyte dialogue was significant and affected microglial cytoskeleton by modulation of cell death and migratory pathways. These were mediated, in part, through F-actin polymerization and filament formation. Astrocyte secretions attenuated HIV-1 infected microglia neurotoxicity and viral growth linked to the regulation of reactive oxygen species. Conclusions These observations provide unique insights into glial crosstalk during disease by supporting astrocyte-mediated regulation of microglial function and its influence on the onset and progression of neuroAIDS. The results open new insights into previously undisclosed pathogenic mechanisms and open the potential for biomarker discovery and therapeutics that may influence the course of HIV-1-mediated neurodegeneration.
Collapse
|
44
|
Yanagita RC, Nakagawa Y, Yamanaka N, Kashiwagi K, Saito N, Irie K. Synthesis, conformational analysis, and biological evaluation of 1-hexylindolactam-V10 as a selective activator for novel protein kinase C isozymes. J Med Chem 2007; 51:46-56. [PMID: 18072722 DOI: 10.1021/jm0706719] [Citation(s) in RCA: 35] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Conventional and novel protein kinase C (PKC) isozymes are the main targets of tumor promoters. We developed 1-hexylindolactam-V10 ( 5) as a selective activator for novel PKC isozymes that play important roles in various cellular processes related to tumor promotion, ischemia--reperfusion injury in the heart, and Alzheimer's disease. The compound existed as a mixture of three conformers. The trans-amide restricted analogues of 5 ( 14 and 15) hardly bound to PKC isozymes, suggesting that the active conformation of 5 could be that with a cis-amide. Compound 5 selectively translocated novel PKC isozymes over conventional PKC isozymes in HeLa cells at 0.1-1 microM. These results suggest that 5 could be useful for the functional analysis of novel PKC isozymes.
Collapse
Affiliation(s)
- Ryo C Yanagita
- Division of Food Science and Biotechnology, Graduate School of Agriculture, Kyoto University, Kyoto 606-8502, Japan
| | | | | | | | | | | |
Collapse
|
45
|
Oyasu M, Fujimiya M, Kashiwagi K, Ohmori S, Imaeda H, Saito N. Immunogold electron microscopic demonstration of distinct submembranous localization of the activated gammaPKC depending on the stimulation. J Histochem Cytochem 2007; 56:253-65. [PMID: 18040079 DOI: 10.1369/jhc.7a7291.2007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
We examined the precise intracellular translocation of gamma subtype of protein kinase C (gammaPKC) after various extracellular stimuli using confocal laser-scanning fluorescent microscopy (CLSM) and immunogold electron microscopy. By CLSM, treatment with 12-O-tetradecanoylphorbol-13-acetate (TPA) resulted in a slow and irreversible accumulation of green fluorescent protein (GFP)-tagged gammaPKC (gammaPKC-GFP) on the plasma membrane. In contrast, treatment with Ca(2+) ionophore and activation of purinergic or NMDA receptors induced a rapid and transient membrane translocation of gammaPKC-GFP. Although each stimulus resulted in PKC localization at the plasma membrane, electron microscopy revealed that gammaPKC showed a subtle but significantly different localization depending on stimulation. Whereas TPA and UTP induced a sustained localization of gammaPKC-GFP on the plasma membrane, Ca(2+) ionophore and NMDA rapidly translocated gammaPKC-GFP to the plasma membrane and then restricted gammaPKC-GFP in submembranous area (<500 nm from the plasma membrane). These results suggest that Ca(2+) influx alone induced the association of gammaPKC with the plasma membrane for only a moment and then located this enzyme at a proper distance in a touch-and-go manner, whereas diacylglycerol or TPA tightly anchored this enzyme on the plasma membrane. The distinct subcellular targeting of gammaPKC in response to various stimuli suggests a novel mechanism for PKC activation.
Collapse
Affiliation(s)
- Miho Oyasu
- Laboratory of Molecular Pharmacology, Biosignal Research Center, Kobe University, Kobe, Japan
| | | | | | | | | | | |
Collapse
|
46
|
Kopitar-Jerala N, Turk B. Cleavage of the myristoylated alanine-rich C kinase substrate (MARCKS) by cysteine cathepsins in cells and tissues of stefin B-deficient mice. Biol Chem 2007; 388:847-52. [PMID: 17655504 DOI: 10.1515/bc.2007.092] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/15/2022]
Abstract
The myristoylated alanine-rich C kinase substrate (MARCKS) is a substrate of protein kinase C (PKC). Besides regulation at the level of gene transcription, MARCKS concentrations within the cell are also regulated by proteolytic cleavage by cathepsins and calpains, which are cysteine proteinases. Stefin B (cystatin B) is an endogenous inhibitor of lysosomal cysteine cathepsins, but not calpains. We have observed increased cleavage of MARCKS in brain and macrophages, but not in liver and kidney extracts of stefin B-deficient mice compared to wild-type mice. Processing of cathepsin B was unaltered in the brain of stefin B-deficient mice and we conclude that increased cleavage of MARCKS could be attributed to the lack of inhibitor.
Collapse
Affiliation(s)
- Natasa Kopitar-Jerala
- Department of Biochemistry, Molecular and Structural Biology, Jozef Stefan Institute, Jamova 39, SI-1000 Ljubljana, Slovenia.
| | | |
Collapse
|
47
|
Abdullah LH, Davis CW. Regulation of airway goblet cell mucin secretion by tyrosine phosphorylation signaling pathways. Am J Physiol Lung Cell Mol Physiol 2007; 293:L591-9. [PMID: 17616647 DOI: 10.1152/ajplung.00150.2007] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mucus hyperproduction in pulmonary obstructive diseases results from increased goblet cell numbers and possibly increased cellular mucin synthesis, occurring in response to inflammatory mediators acting via receptor tyrosine kinases (RYK) and tyrosine phosphorylation (Y-Pi) signaling pathways. Yet, increased mucin synthesis does not lead necessarily to increased secretion, as mucins are stored in secretory granules and secreted in response to extracellular signals, commonly assumed to be mediated by G protein-coupled receptors (GPCRs). We asked whether activation 1) of Y-Pi signaling pathways, in principal, and 2) of the novel PKC isoform, nPKCdelta, by Y-Pi, specifically, might lead to regulated mucin secretion. nPKCdelta in SPOC1 cells was tyrosine phosphorylated by exposure to purinergic agonist (ATPgammaS) or PMA, actions that were blocked by the Src kinase inhibitor, PP1. Mucin secretion, however, was not affected by PP1. Hence, activation of nPKCdelta by Y-Pi is unlikely to participate in GPCR-related mucin secretion. Mucin secretion from both SPOC1 and normal human bronchial epithelial (NHBE) cells was stimulated by generalized protein Y-Pi induced by the tyrosine phosphatase inhibitor, pervanadate (PV). PV-induced SPOC1 cell mucin secretion was not affected by inhibition of Src kinases (genistein or PP1), or of PI3 kinase (LY-294002). MAP kinase pathway inhibitors, RAF1 kinase inhibitor-I and U0126 (MEK), inhibited SPOC1 cell PV-induced secretion by approximately 50%. Significantly, the phospholipase C (PLC) inhibitor, U-73122, essentially abolished PV- and ATPgammaS-induced mucin secretion from both SPOC1 and NHBE cells. Hence, PLC signaling may play a key role in regulated mucin secretion, whether the event is initiated by mediators interacting with GPCRs or RYKs.
Collapse
Affiliation(s)
- Lubna H Abdullah
- Cystic Fibrosis/Pulmonary Research and Treatment Center, University of North Carolina, Chapel Hill, North Carolina 27599, USA
| | | |
Collapse
|
48
|
Salcedo M, Cuevas C, Alonso JL, Otero G, Faircloth G, Fernandez-Sousa JM, Avila J, Wandosell F. The marine sphingolipid-derived compound ES 285 triggers an atypical cell death pathway. Apoptosis 2007; 12:395-409. [PMID: 17191124 DOI: 10.1007/s10495-006-0573-z] [Citation(s) in RCA: 45] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
The isolation of new molecules from marine sources opens the door to their possible therapeutic use against tumors and other pathological conditions. Indeed, we recently defined the cytotoxicity of ES 285, obtained from the clam Mactromeris polynima, and its affects on the cells microfilament but not the microtubule network. Considering the analogy between ES 285 and sphingosine-related lipids, we wondered whether ES 285 might affect the activity of PKC at the intracellular level. While we anticipated that ES 285 might inhibit PKC, it turns out that in contrast it serves to activate PKC at the cellular level. Indeed, like other sphingosine-related lipids, ES 285 induces the phosphorylation of MARCKS. Additionally, we further examined the cytotoxicity of ES 285 to elucidate the molecular mechanisms through which this compound triggers apoptosis. When the influence of ES 285 on "cell death markers" was assessed, it became clear that ES285 activates caspase 3 and 12, and that it modified the phosphorylation of p53. In contrast, ES 285 does not affect other pathways widely implicated in regulating cell survival/apoptosis, such as JNK, Erks or Akt. Thus, these data suggest that ES 285-triggers an atypical cell death program when compared to other sphingosine-dependent apoptosis pathways.
Collapse
Affiliation(s)
- M Salcedo
- Centro de Biología Molecular Severo Ochoa, Facultad de Ciencias, Universidad Autónoma de Madrid, 28049, Madrid, Spain
| | | | | | | | | | | | | | | |
Collapse
|
49
|
Tsuruno S, Hirano T. Persistent activation of protein kinase Calpha is not necessary for expression of cerebellar long-term depression. Mol Cell Neurosci 2007; 35:38-48. [PMID: 17363267 DOI: 10.1016/j.mcn.2007.01.016] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2006] [Revised: 01/05/2007] [Accepted: 01/30/2007] [Indexed: 10/23/2022] Open
Abstract
Protein kinase Calpha (PKCalpha) plays a major role in the induction of long-term depression (LTD) in a cerebellar Purkinje cell (PC). The sequential activation model for classical PKC states that PKCalpha translocates to the plasma membrane by binding Ca(++) and then becomes fully activated by binding diacylglycerol (DAG), which enables estimation of the activity by monitoring its localization. Here, we performed simultaneous electrophysiological recording and fluorescence imaging in a cultured PC expressing GFP-tagged PKCalpha. When a PC was depolarized, PKCalpha transiently translocated to the plasma membrane in a Ca(++)-dependent manner. Application of membrane permeable DAG or the blocker of DAG lipase prolonged the translocation. These results suggest that the sequential activation model is applicable to PCs. Conjunctive applications of glutamate and depolarization pulse induced LTD, but did not prolong the translocation. Thus, our results imply that persistent activation of PKCalpha is not necessary for the expression of LTD.
Collapse
Affiliation(s)
- S Tsuruno
- Department of Biophysics, Graduate School of Science, Kyoto University, Kyoto 606-8502, Japan
| | | |
Collapse
|
50
|
Shiraishi M, Tanabe A, Saito N, Sasaki Y. Unphosphorylated MARCKS is involved in neurite initiation induced by insulin-like growth factor-I in SH-SY5Y cells. J Cell Physiol 2007; 209:1029-38. [PMID: 16941482 DOI: 10.1002/jcp.20814] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Myristoylated alanine-rich C kinase substrate (MARCKS) has been suggested to be involved in various aspects of neuronal cell differentiation, including neurite outgrowth. However, the precise mechanisms by which MARCKS phosphorylation is regulated, and how MARCKS contributes to neurite outgrowth, are poorly understood. Here, we found that treatment of SH-SY5Y cells with insulin-like growth factor-I (IGF-I) induced a rapid and transient decrease in the level of phosphorylated MARCKS (P-MARCKS) to below the basal level. The decrease in P-MARCKS induced by IGF-I was blocked by pretreatment of cells with phosphoinositide 3-kinase (PI3K) inhibitors, LY294002 and wortmannin. A decrease in P-MARCKS was also observed in cells treated with a Rho-dependent kinase (ROCK) inhibitor, Y27632. Furthermore, IGF-I induced transient inactivation of RhoA, an upstream effector of ROCK. We showed that MARCKS was translocated to the membrane and colocalized with F-actin at the lamellipodia and the tips of neurites in the cells stimulated with IGF-I. Finally, overexpression of wild-type MARCKS or an unphosphorylatable mutant of MARCKS enhanced the number of neurite-bearing cells relative to vector-transfected cells. Taken together, these findings suggest that unphosphorylated MARCKS is involved in neurite initiation, and highlight the important role played by MARCKS in organization of the actin cytoskeleton.
Collapse
Affiliation(s)
- Mitsuya Shiraishi
- Laboratory of Pharmacology, School of Pharmaceutical Science, Kitasato University, Shirokane 5-9-1, Minato-ku, Tokyo 108-8641, Japan.
| | | | | | | |
Collapse
|