1
|
Jeon HM, Noh HS, Jeon MG, Park JH, Lee YS, Seo G, Cheon YH, Kim M, Han MK, Park JY, Lee SI. The HRAS-binding C2 domain of PLCη2 suppresses tumor-like synoviocytes and experimental arthritis in rheumatoid arthritis. Exp Mol Med 2025; 57:335-348. [PMID: 39894825 PMCID: PMC11873285 DOI: 10.1038/s12276-025-01393-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2024] [Revised: 11/08/2024] [Accepted: 11/12/2024] [Indexed: 02/04/2025] Open
Abstract
Fibroblast-like synoviocytes (FLSs), which are stromal cells that play key roles in rheumatoid arthritis (RA) pathophysiology, are characterized by a tumor-like phenotype and immunostimulatory actions. C2 domains in various proteins play roles in intracellular signaling and altering cellular characteristics, and some C2 domain-containing proteins exacerbate or alleviate certain malignant or inflammatory diseases. However, the roles of C2 domains in regulating the functions of RA FLSs remain unclear. Here we performed functional C2 domainomics with 144 C2 domain-containing viral vectors and identified the C2 domain of PLCη2 as a key regulator of RA FLSs. In mice, overexpressing PLCη2 or only its C2 domain PLCη2 (PLCη2_C2) diminished the proliferation, migration, invasion and inflammatory responses of RA FLSs, mitigating RA pathology; the absence of PLCη2 amplified these proinflammatory and destructive processes in RA FLSs in vivo. Mechanistically, PLCη2 and PLCη2_C2 participate in the pathological signaling of RA FLSs in a calcium-independent manner through protein-protein interactions. Specifically, PLCη2_C2 disrupted HRAS-RAF1 interactions, suppressing downstream signaling pathways, including the NF-κB, JAK-STAT and MAPK pathways. Collectively, these findings establish PLCη2 and PLCη2_C2 as novel inhibitory regulators in RA, suggesting promising therapeutic avenues for addressing FLS-driven disease mechanisms.
Collapse
Affiliation(s)
- Hyun Min Jeon
- Division of Rheumatology, Department of Internal Medicine and Institute of Medical Science, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, South Korea
| | - Hae Sook Noh
- Division of Rheumatology, Department of Internal Medicine and Institute of Medical Science, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, South Korea
| | - Min-Gyu Jeon
- Division of Rheumatology, Department of Internal Medicine and Institute of Medical Science, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, South Korea
| | - Jin-Ho Park
- Division of Rheumatology, Department of Internal Medicine and Institute of Medical Science, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, South Korea
| | - Young-Sun Lee
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, South Korea
| | - Gyunghwa Seo
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, South Korea
| | - Yun-Hong Cheon
- Division of Rheumatology, Department of Internal Medicine and Institute of Medical Science, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, South Korea
| | - Mingyo Kim
- Division of Rheumatology, Department of Internal Medicine and Institute of Medical Science, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, South Korea
| | - Myung-Kwan Han
- Department of Microbiology, Jeonbuk National University Medical School, Jeonju, South Korea
| | - Jae-Yong Park
- School of Biosystem and Biomedical Science, College of Health Science, Korea University, Seoul, South Korea.
| | - Sang-Il Lee
- Division of Rheumatology, Department of Internal Medicine and Institute of Medical Science, Gyeongsang National University School of Medicine and Gyeongsang National University Hospital, Jinju, South Korea.
| |
Collapse
|
2
|
Šafranek M, Shumbusho A, Johansen W, Šarkanová J, Voško S, Bokor B, Jásik J, Demko V. Membrane-anchored calpains - hidden regulators of growth and development beyond plants? FRONTIERS IN PLANT SCIENCE 2023; 14:1289785. [PMID: 38173928 PMCID: PMC10762896 DOI: 10.3389/fpls.2023.1289785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 09/06/2023] [Accepted: 12/04/2023] [Indexed: 01/05/2024]
Abstract
Calpains are modulatory proteases that modify diverse cellular substrates and play essential roles in eukaryots. The best studied are animal cytosolic calpains. Here, we focus on enigmatic membrane-anchored calpains, their structural and functional features as well as phylogenetic distribution. Based on domain composition, we identified four types of membrane-anchored calpains. Type 1 and 2 show broad phylogenetic distribution among unicellular protists and streptophytes suggesting their ancient evolutionary origin. Type 3 and 4 diversified early and are present in brown algae and oomycetes. The plant DEK1 protein is the only representative of membrane-anchored calpains that has been functionally studied. Here, we present up to date knowledge about its structural features, putative regulation, posttranslational modifications, and biological role. Finally, we discuss potential model organisms and available tools for functional studies of membrane-anchored calpains with yet unknown biological role. Mechanistic understanding of membrane-anchored calpains may provide important insights into fundamental principles of cell polarization, cell fate control, and morphogenesis beyond plants.
Collapse
Affiliation(s)
- Martin Šafranek
- Institute of Botany, Plant Science and Biodiversity Centre, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Alain Shumbusho
- Department of Plant Physiology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia
| | - Wenche Johansen
- Faculty of Applied Ecology, Agricultural Sciences and Biotechnology, Inland Norway University of Applied Sciences, Hamar, Norway
| | - Júlia Šarkanová
- Comenius University Science Park, Comenius University in Bratislava, Bratislava, Slovakia
| | - Stanislav Voško
- Comenius University Science Park, Comenius University in Bratislava, Bratislava, Slovakia
| | - Boris Bokor
- Department of Plant Physiology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia
- Comenius University Science Park, Comenius University in Bratislava, Bratislava, Slovakia
| | - Ján Jásik
- Institute of Botany, Plant Science and Biodiversity Centre, Slovak Academy of Sciences, Bratislava, Slovakia
| | - Viktor Demko
- Institute of Botany, Plant Science and Biodiversity Centre, Slovak Academy of Sciences, Bratislava, Slovakia
- Department of Plant Physiology, Faculty of Natural Sciences, Comenius University in Bratislava, Bratislava, Slovakia
| |
Collapse
|
3
|
Lou YX, Gu J, Zhu L, Sun SQ, Hao XL, Chen JP, Han F, Wang DD, Jiang X, Liu JY. TC2N Promotes Cell Proliferation and Metastasis in Hepatocellular Carcinoma by Targeting the Wnt/β-Catenin Signaling Pathway. J Transl Med 2023; 103:100260. [PMID: 37839635 DOI: 10.1016/j.labinv.2023.100260] [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: 02/14/2023] [Revised: 09/14/2023] [Accepted: 10/07/2023] [Indexed: 10/17/2023] Open
Abstract
Hepatocellular carcinoma (HCC), one of the most prevalent types of cancer worldwide, has an exceedingly poor prognosis. Tandem C2 domain nuclear protein (TC2N) has been implicated in tumorigenesis and serves as an oncogene or tumor suppressor in different types of cancer. Here, we explore the possible regulatory activities and molecular mechanisms of TC2N in HCC progression. However, TC2N expression was significantly upregulated in HCC tissues and hepatoma cell lines, and this upregulation was positively correlated with tumor progression in HCC patients. The ectopic overexpression of TC2N accelerated the proliferation, migration, and invasion of HCC cells, whereas its knockdown showed the opposite effects. Bioinformatics analysis showed that TC2N participates in the regulation of the Wnt/β-catenin signaling pathway. Mechanistically, TC2N activated the Wnt/β-catenin signaling pathway by regulating the expression levels of β-catenin and its downstream targets CyclinD1, MMP7, c-Myc, c-Jun, AXIN2, and glutamine synthase. Furthermore, the deletion of β-catenin effectively neutralized the regulation of TC2N in HCC proliferation and metastasis. Overall, this study showed that TC2N promotes HCC proliferation and metastasis by activating the Wnt/β-catenin signaling pathway, indicating that TC2N might be a potential molecular target for the treatment of HCC.
Collapse
Affiliation(s)
- Yi-Xia Lou
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jing Gu
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Lei Zhu
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Sheng-Qi Sun
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Xiang-Lin Hao
- Department of Pathology, Xinqiao Hospital, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jian-Ping Chen
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Fei Han
- Department of Toxicology, School of Public Health, Chongqing Medical University, Chongqing, China
| | - Dan-Dan Wang
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, China; Laboratory of Cell Signal Transduction, Henan Provincial Engineering Centre for Tumor Molecular Medicine, School of Basic Medicine, Henan University, Kaifeng, China
| | - Xiao Jiang
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, China
| | - Jin-Yi Liu
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University (Army Medical University), Chongqing, China.
| |
Collapse
|
4
|
Xu Y, Tao J, Hou Y, Zhang S. Effects of PKCε knockdown on mitochondrial membrane potential of human glioma cells in vitro and growth of U251 cell-derived tumors in vivo. Biotech Histochem 2023; 98:501-507. [PMID: 37501577 DOI: 10.1080/10520295.2023.2240226] [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] [Indexed: 07/29/2023] Open
Abstract
Glioma is the most common type of primary brain tumor; it exhibits great invasive capacity, morbidity and mortality. Protein kinase Cε (PKCε), a serine/threonine kinase, contributes to the development and progression of many cancers. We investigated whether knockdown of PKCε could affect the mitochondrial membrane potential of human glioma cell lines, U251 and U87, and the growth of U251 cell-derived tumors in nude mice. We found that the expression of PKCε was greater in human glioma tissues than in human normal brain tissues. Knockdown of PKCε reduced mitochondrial membrane potential in U251 and U87 cells. Knockdown of PKCε also suppressed the growth of tumors derived from U251 cells and induced apoptosis of U251 cells in vivo. Our findings indicate that PKCε is important for development and progression of glioma and may be a potential therapeutic target for glioma treatment.
Collapse
Affiliation(s)
- Yaming Xu
- School of Medicine, Shanghai University, Shanghai, P. R. China
| | - Jie Tao
- Central Laboratory, Department of Neurology, Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| | - Yafei Hou
- Shanghai Institute of Immunology, Shanghai Jiao Tong University School of Medicine, Shanghai, P. R. China
| | - Shuzhang Zhang
- Central Laboratory, Department of Neurology, Interventional Cancer Institute of Chinese Integrative Medicine, Putuo Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, P. R. China
| |
Collapse
|
5
|
Wang D, Zhao Z, Long Y, Fan R. Protein Kinase C Is Involved in Vegetative Development, Stress Response and Pathogenicity in Verticillium dahliae. Int J Mol Sci 2023; 24:14266. [PMID: 37762573 PMCID: PMC10531995 DOI: 10.3390/ijms241814266] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/31/2023] [Revised: 08/30/2023] [Accepted: 09/04/2023] [Indexed: 09/29/2023] Open
Abstract
Potato Verticillium wilt, caused by Verticillium dahliae, is a serious soil-borne vascular disease, which restricts the sustainable development of the potato industry, and the pathogenic mechanism of the fungus is complex. Therefore, it is of great significance to explore the important pathogenic factors of V. dahliae to expand the understanding of its pathology. Protein kinase C (PKC) gene is located in the Ca2+ signaling pathway, which is highly conserved in filamentous fungi and involved in the regulation of a variety of biological processes. In the current study, the PKC gene in V. dahliae (VdPKC) was characterized, and its effects on the fungal pathogenicity and tolerance to fungicide stress were further studied. The results showed that the VdPKC positively regulated the growth and development, conidial germination, and production of V. dahliae, which was necessary for the fungus to achieve pathogenicity. It also affected the formation of melanin and microsclerotia and changed the adaptability of V. dahliae to different environmental stresses. In addition, VdPKC altered the tolerance of V. dahliae to different fungicides, which may be a potential target for polyoxin. Therefore, our results strongly suggest that VdPKC gene is necessary for the vegetative growth, stress response, and pathogenicity of V. dahliae.
Collapse
Affiliation(s)
| | | | | | - Rong Fan
- College of Agriculture, Guizhou University, Guiyang 550025, China; (D.W.); (Z.Z.); (Y.L.)
| |
Collapse
|
6
|
Cai YJ, He JY, Yang XY, Huang W, Fu XM, Guo SQ, Yang JJ, Dong JD, Zeng HT, Wu YJ, Qin Z, Qin QW, Sun HY. Molecular characterization, expression and function analysis of Epinephelus coioides PKC-ɑ response to Singapore grouper iridovirus (SGIV) infection. DEVELOPMENTAL AND COMPARATIVE IMMUNOLOGY 2023; 142:104646. [PMID: 36702214 DOI: 10.1016/j.dci.2023.104646] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/16/2022] [Revised: 12/28/2022] [Accepted: 01/20/2023] [Indexed: 06/18/2023]
Abstract
Protein kinase C (PKC) constitutes the main signal transduction pathway, and participates in the signal pathway of cell proliferation and movement in mammals. In this study, PKC-ɑ was obtained from Epinephelus coioides, an important marine fish cultivated in the coastal areas of southern China and Southeast Asia. The full length cDNA of PKC-ɑ was 3362 bp in length containing a 23 bp 5'UTR, a 1719 bp 3'UTR, and a 1620 bp open reading frame encoding 539 amino acids. It contains three conservative domains including protein kinase C conserved region 2 (C2), Serine/Threonine protein kinases, catalytic domain (S_TKc) and ser/thr-type protein kinases (S_TK_X). Its mRNA can be detected in all 11 tissues examined of E. coioides, and the expression was significantly upregulated response to Singapore grouper iridovirus (SGIV) infection, one of the important pathogens of marine fish. Upregulated E. coioides PKC-ɑ significantly inhibited the activation of nuclear factor kappa-B (NF-κB) and activator protein-1 (AP-1), and SGIV-induced cell apoptosis. The results indicated that the PKC-ɑ may play an important role in pathogenic stimulation.
Collapse
Affiliation(s)
- Yi-Jie Cai
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Jia-Yang He
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Xin-Yue Yang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Wei Huang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Xue-Mei Fu
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Shi-Qing Guo
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Jie-Jia Yang
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Jun-De Dong
- Guangdong Provincial Key Laboratory of Applied Marine Biology, 510301, PR China
| | - Hai-Tian Zeng
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Yan-Jun Wu
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Zhou Qin
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China
| | - Qi-Wei Qin
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China; Southern Marine Science and Engineering Guangdong Laboratory, Zhuhai, 519000, PR China; Laboratory for Marine Biology and Biotechnology, Qingdao National Laboratory for Marine Science and Technology, Qingdao, 266000, PR China.
| | - Hong-Yan Sun
- Guangdong Laboratory for Lingnan Modern Agriculture, College of Marine Sciences, South China Agricultural University, Guangzhou, 510642, Guangdong Province, PR China.
| |
Collapse
|
7
|
Paeonol protects against doxorubicin-induced cardiotoxicity by promoting Mfn2-mediated mitochondrial fusion through activating the PKCε-Stat3 pathway. J Adv Res 2022; 47:151-162. [PMID: 35842187 PMCID: PMC10173194 DOI: 10.1016/j.jare.2022.07.002] [Citation(s) in RCA: 33] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/22/2022] [Revised: 06/13/2022] [Accepted: 07/10/2022] [Indexed: 11/21/2022] Open
Abstract
INTRODUCTION The anti-cancer medication doxorubicin (Dox) is largely restricted in clinical usage due to its significant cardiotoxicity. The only medication approved by the FDA for Dox-induced cardiotoxicity is dexrazoxane, while it may reduce the sensitivity of cancer cells to chemotherapy and is restricted for use. There is an urgent need for the development of safe and effective medicines to alleviate Dox-induced cardiotoxicity. OBJECTIVES The objective of this study was to determine whether Paeonol (Pae) has the ability to protect against Dox-induced cardiotoxicity and if so, what are the underlying mechanisms involved. METHODS Sprague-Dawley rats and primary cardiomyocytes were used to create Dox-induced cardiotoxicity models. Pae's effects on myocardial damage, mitochondrial function, mitochondrial dynamics and signaling pathways were studied using a range of experimental methods. RESULTS Pae enhanced Mfn2-mediated mitochondrial fusion, restored mitochondrial function and cardiac performance both in vivo and in vitro under the Dox conditions. The protective properties of Pae were blunted when Mfn2 was knocked down or knocked out in Dox-induced cardiomyocytes and hearts respectively. Mechanistically, Pae promoted Mfn2-mediated mitochondria fusion by activating the transcription factor Stat3, which bound to the Mfn2 promoter in a direct manner and up-regulated its transcriptional expression. Furthermore, molecular docking, surface plasmon resonance and co-immunoprecipitation studies showed that Pae's direct target was PKCε, which interacted with Stat3 and enabled its phosphorylation and activation. Pae-induced Stat3 phosphorylation and Mfn2-mediated mitochondrial fusion were inhibited when PKCε was knocked down. Furthermore, Pae did not interfere with Dox's antitumor efficacy in several tumor cells. CONCLUSION Pae protects the heart against Dox-induced damage by stimulating mitochondrial fusion via the PKCε-Stat3-Mfn2 pathway, indicating that Pae might be a promising therapeutic therapy for Dox-induced cardiotoxicity while maintaining Dox's anticancer activity.
Collapse
|
8
|
Black JD, Affandi T, Black AR, Reyland ME. PKCα and PKCδ: Friends and Rivals. J Biol Chem 2022; 298:102194. [PMID: 35760100 PMCID: PMC9352922 DOI: 10.1016/j.jbc.2022.102194] [Citation(s) in RCA: 14] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/10/2022] [Revised: 06/13/2022] [Accepted: 06/21/2022] [Indexed: 01/06/2023] Open
Abstract
PKC comprises a large family of serine/threonine kinases that share a requirement for allosteric activation by lipids. While PKC isoforms have significant homology, functional divergence is evident among subfamilies and between individual PKC isoforms within a subfamily. Here, we highlight these differences by comparing the regulation and function of representative PKC isoforms from the conventional (PKCα) and novel (PKCδ) subfamilies. We discuss how unique structural features of PKCα and PKCδ underlie differences in activation and highlight the similar, divergent, and even opposing biological functions of these kinases. We also consider how PKCα and PKCδ can contribute to pathophysiological conditions and discuss challenges to targeting these kinases therapeutically.
Collapse
Affiliation(s)
- Jennifer D Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE.
| | - Trisiani Affandi
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus
| | - Adrian R Black
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, Omaha, NE
| | - Mary E Reyland
- Department of Craniofacial Biology, School of Dental Medicine, University of Colorado Anschutz Medical Campus.
| |
Collapse
|
9
|
Global Identification and Characterization of C2 Domain-Containing Proteins Associated with Abiotic Stress Response in Rice (Oryza sativa L.). Int J Mol Sci 2022; 23:ijms23042221. [PMID: 35216337 PMCID: PMC8875736 DOI: 10.3390/ijms23042221] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/28/2022] [Revised: 02/14/2022] [Accepted: 02/15/2022] [Indexed: 11/20/2022] Open
Abstract
C2 domain-containing proteins (C2DPs) have been identified in different genomes that contain single or multiple C2 domains in their C- or N-terminal. It possesses higher functional activity in the transmembrane regions. The identification of C2 domains were reported in a previous study, such as multiple C2 domains and transmembrane-region proteins (MCTPs) and N-terminal-TM-C2 domain proteins (NTMC2s) of rice, Arabidopsis thaliana, and cotton, whereas the C2DP gene family in rice has not been comprehensively studied, and the role of the C2DP gene in rice in response to abiotic stress is not yet fully understood. In this study, we identified 82 C2DPs in the rice genome and divided them into seven groups through phylogenetic analysis. The synteny analysis revealed that duplication events were either exhibited within the genome of rice or between the genomes of rice and other species. Through the analysis of cis-acting elements in promoters, expression profiles, and qRT-PCR results, the functions of OsC2DPs were found to be widely distributed in diverse tissues and were extensively involved in phytohormones-related and abiotic stresses response in rice. The prediction of the microRNA (miRNA) targets of OsC2DPs revealed the possibility of regulation by consistent miRNAs. Notably, OsC2DP50/51/52 as a co-tandem duplication exhibited similar expression variations and involved the coincident miRNA-regulation pathway. Moreover, the results of the genotypic variation and haplotype analysis revealed that OsC2DP17, OsC2DP29, and OsC2DP49 were associated with cold stress responses. These findings provided comprehensive insights for characterizations of OsC2DPs in rice as well as for their roles for abiotic stress.
Collapse
|
10
|
Trop-2, Na+/K+ ATPase, CD9, PKCα, cofilin assemble a membrane signaling super-complex that drives colorectal cancer growth and invasion. Oncogene 2022; 41:1795-1808. [DOI: 10.1038/s41388-022-02220-1] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2021] [Revised: 01/10/2022] [Accepted: 01/27/2022] [Indexed: 12/20/2022]
|
11
|
The redundancy and diversity between two novel PKC isotypes that regulate learning in Caenorhabditis elegans. Proc Natl Acad Sci U S A 2022; 119:2106974119. [PMID: 35027448 PMCID: PMC8784152 DOI: 10.1073/pnas.2106974119] [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] [Accepted: 11/16/2021] [Indexed: 11/18/2022] Open
Abstract
The nematode Caenorhabditis elegans learns the concentration of NaCl and moves toward the previously experienced concentration. In this behavior, the history of NaCl concentration change is reflected in the level of diacylglycerol and the activity of protein kinase C, PKC-1, in the gustatory sensory neuron ASER and determines the direction of migration. Here, through a genetic screen, we found that the activation of Gq protein compensates for the behavioral defect of the loss-of-function mutant of pkc-1 We found that Gq activation results in hyperproduction of diacylglycerol in ASER sensory neuron, which leads to recruitment of TPA-1, an nPKC isotype closely related to PKC-1. Unlike the pkc-1 mutants, loss of tpa-1 did not obviously affect migration directions in the conventional learning assay. This difference was suggested to be due to cooperative functions of the C1 and C2-like domains of the nPKC isotypes. Furthermore, we investigated how the compensatory capability of tpa-1 contributes to learning and found that learning was less robust in the context of cognitive decline or environmental perturbation in tpa-1 mutants. These results highlight how two nPKC isotypes contribute to the learning system.
Collapse
|
12
|
Jan CR, Hao LJ, Lin RA, Chen LC, Wang JL, Chen IS, Kuo CC, Chou CT, Chien JM. Action of the natural compound gomisin a on Ca 2+ movement in human prostate cancer cells. CHINESE J PHYSIOL 2022; 65:151-157. [DOI: 10.4103/cjp.cjp_6_22] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
13
|
Li H, Fang H, Chang L, Qiu S, Ren X, Cao L, Bian J, Wang Z, Guo Y, Lv J, Sun Z, Wang T, Li B. TC2N: A Novel Vital Oncogene or Tumor Suppressor Gene In Cancers. Front Immunol 2021; 12:764749. [PMID: 34925334 PMCID: PMC8674203 DOI: 10.3389/fimmu.2021.764749] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/25/2021] [Accepted: 10/29/2021] [Indexed: 12/12/2022] Open
Abstract
Several C2 domain-containing proteins play key roles in tumorigenesis, signal transduction, and mediating protein–protein interactions. Tandem C2 domains nuclear protein (TC2N) is a tandem C2 domain-containing protein that is differentially expressed in several types of cancers and is closely associated with tumorigenesis and tumor progression. Notably, TC2N has been identified as an oncogene in lung and gastric cancer but as a tumor suppressor gene in breast cancer. Recently, a large number of tumor-associated antigens (TAAs), such as heat shock proteins, alpha-fetoprotein, and carcinoembryonic antigen, have been identified in a variety of malignant tumors. Differences in the expression levels of TAAs between cancer cells and normal cells have led to these antigens being investigated as diagnostic and prognostic biomarkers and as novel targets in cancer treatment. In this review, we summarize the clinical characteristics of TC2N-positive cancers and potential mechanisms of action of TC2N in the occurrence and development of specific cancers. This article provides an exploration of TC2N as a potential target for the diagnosis and treatment of different types of cancers.
Collapse
Affiliation(s)
- Hanyang Li
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
- Department of Thyroid Surgery, The Second Hospital of Jilin University, Changchun, China
| | - He Fang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Li Chang
- Department of Pathology, The Second Hospital of Jilin University, Changchun, China
| | - Shuang Qiu
- Department of Biobank, The China-Japan Union Hospital of Jilin University, Changchun, China
| | - Xiaojun Ren
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
| | - Lidong Cao
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Jinda Bian
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Zhenxiao Wang
- Department of Hepatobiliary and Pancreatic Surgery, The Second Hospital of Jilin University, Changchun, China
| | - Yi Guo
- Department of Breast Surgery, The Affiliated Hospital Changchun University of Chinese Medicine, Changchun, China
| | - Jiayin Lv
- Department of Orthopedics, The China-Japan Union Hospital of Jilin University, Changchun, China
| | - Zhihui Sun
- Department of Pharmacy, The Second Hospital of Jilin University, Changchun, China
| | - Tiejun Wang
- Department of Radiotherapy, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Tiejun Wang, ; Bingjin Li,
| | - Bingjin Li
- Jilin Provincial Key Laboratory on Molecular and Chemical Genetic, The Second Hospital of Jilin University, Changchun, China
- *Correspondence: Tiejun Wang, ; Bingjin Li,
| |
Collapse
|
14
|
Khan SM, Faisal ARM, Nila TA, Binti NN, Hosen MI, Shekhar HU. A computational in silico approach to predict high-risk coding and non-coding SNPs of human PLCG1 gene. PLoS One 2021; 16:e0260054. [PMID: 34793541 PMCID: PMC8601573 DOI: 10.1371/journal.pone.0260054] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2021] [Accepted: 10/31/2021] [Indexed: 11/28/2022] Open
Abstract
PLCG1 gene is responsible for many T-cell lymphoma subtypes, including peripheral T-cell lymphoma (PTCL), angioimmunoblastic T-cell lymphoma (AITL), cutaneous T-cell lymphoma (CTCL), adult T-cell leukemia/lymphoma along with other diseases. Missense mutations of this gene have already been found in patients of CTCL and AITL. The non-synonymous single nucleotide polymorphisms (nsSNPs) can alter the protein structure as well as its functions. In this study, probable deleterious and disease-related nsSNPs in PLCG1 were identified using SIFT, PROVEAN, PolyPhen-2, PhD-SNP, Pmut, and SNPS&GO tools. Further, their effect on protein stability was checked along with conservation and solvent accessibility analysis by I-mutant 2.0, MUpro, Consurf, and Netsurf 2.0 server. Some SNPs were finalized for structural analysis with PyMol and BIOVIA discovery studio visualizer. Out of the 16 nsSNPs which were found to be deleterious, ten nsSNPs had an effect on protein stability, and six mutations (L411P, R355C, G493D, R1158H, A401V and L455F) were predicted to be highly conserved. Among the six highly conserved mutations, four nsSNPs (R355C, A401V, L411P and L455F) were part of the catalytic domain. L411P, L455F and G493D made significant structural change in the protein structure. Two mutations-Y210C and R1158H had post-translational modification. In the 5' and 3' untranslated region, three SNPs, rs139043247, rs543804707, and rs62621919 showed possible miRNA target sites and DNA binding sites. This in silico analysis has provided a structured dataset of PLCG1 gene for further in vivo researches. With the limitation of computational study, it can still prove to be an asset for the identification and treatment of multiple diseases associated with the target gene.
Collapse
Affiliation(s)
- Safayat Mahmud Khan
- Department of Biochemistry and Molecular Biology, Clinical Biochemistry and Translational Medicine Laboratory, University of Dhaka, Dhaka, Bangladesh
| | - Ar-Rafi Md. Faisal
- Department of Biochemistry and Molecular Biology, Clinical Biochemistry and Translational Medicine Laboratory, University of Dhaka, Dhaka, Bangladesh
| | - Tasnin Akter Nila
- Department of Biochemistry and Molecular Biology, Clinical Biochemistry and Translational Medicine Laboratory, University of Dhaka, Dhaka, Bangladesh
| | - Nabila Nawar Binti
- Department of Biochemistry and Molecular Biology, Clinical Biochemistry and Translational Medicine Laboratory, University of Dhaka, Dhaka, Bangladesh
| | - Md. Ismail Hosen
- Department of Biochemistry and Molecular Biology, Clinical Biochemistry and Translational Medicine Laboratory, University of Dhaka, Dhaka, Bangladesh
| | - Hossain Uddin Shekhar
- Department of Biochemistry and Molecular Biology, Clinical Biochemistry and Translational Medicine Laboratory, University of Dhaka, Dhaka, Bangladesh
| |
Collapse
|
15
|
Activators and Inhibitors of Protein Kinase C (PKC): Their Applications in Clinical Trials. Pharmaceutics 2021; 13:pharmaceutics13111748. [PMID: 34834162 PMCID: PMC8621927 DOI: 10.3390/pharmaceutics13111748] [Citation(s) in RCA: 60] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/08/2021] [Revised: 10/18/2021] [Accepted: 10/19/2021] [Indexed: 02/05/2023] Open
Abstract
Protein kinase C (PKC), a family of phospholipid-dependent serine/threonine kinase, is classed into three subfamilies based on their structural and activation characteristics: conventional or classic PKC isozymes (cPKCs; α, βI, βII, and γ), novel or non-classic PKC isozymes (nPKCs; δ, ε, η, and θ), and atypical PKC isozymes (aPKCs; ζ, ι, and λ). PKC inhibitors and activators are used to understand PKC-mediated intracellular signaling pathways and for the diagnosis and treatment of various PKC-associated diseases, such as cancers, neurological diseases, cardiovascular diseases, and infections. Many clinical trials of PKC inhibitors in cancers showed no significant clinical benefits, meaning that there is a limitation to design a cancer therapeutic strategy targeting PKC alone. This review will focus on the activators and inhibitors of PKC and their applications in clinical trials.
Collapse
|
16
|
Andronis CE, Hane JK, Bringans S, Hardy GESJ, Jacques S, Lipscombe R, Tan KC. Gene Validation and Remodelling Using Proteogenomics of Phytophthora cinnamomi, the Causal Agent of Dieback. Front Microbiol 2021; 12:665396. [PMID: 34394023 PMCID: PMC8360494 DOI: 10.3389/fmicb.2021.665396] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/08/2021] [Accepted: 05/18/2021] [Indexed: 11/13/2022] Open
Abstract
Phytophthora cinnamomi is a pathogenic oomycete that causes plant dieback disease across a range of natural ecosystems and in many agriculturally important crops on a global scale. An annotated draft genome sequence is publicly available (JGI Mycocosm) and suggests 26,131 gene models. In this study, soluble mycelial, extracellular (secretome), and zoospore proteins of P. cinnamomi were exploited to refine the genome by correcting gene annotations and discovering novel genes. By implementing the diverse set of sub-proteomes into a generated proteogenomics pipeline, we were able to improve the P. cinnamomi genome annotation. Liquid chromatography mass spectrometry was used to obtain high confidence peptides with spectral matching to both the annotated genome and a generated 6-frame translation. Two thousand seven hundred sixty-four annotations from the draft genome were confirmed by spectral matching. Using a proteogenomic pipeline, mass spectra were used to edit the P. cinnamomi genome and allowed identification of 23 new gene models and 60 edited gene features using high confidence peptides obtained by mass spectrometry, suggesting a rate of incorrect annotations of 3% of the detectable proteome. The novel features were further validated by total peptide support, alongside functional analysis including the use of Gene Ontology and functional domain identification. We demonstrated the use of spectral data in combination with our proteogenomics pipeline can be used to improve the genome annotation of important plant diseases and identify missed genes. This study presents the first use of spectral data to edit and manually annotate an oomycete pathogen.
Collapse
Affiliation(s)
- Christina E Andronis
- Centre for Crop and Disease Management, Curtin University, Bentley, WA, Australia.,Proteomics International, Nedlands, WA, Australia
| | - James K Hane
- Centre for Crop and Disease Management, Curtin University, Bentley, WA, Australia.,Faculty of Science and Engineering, Curtin Institute for Computation, Curtin University, Perth, WA, Australia
| | | | - Giles E S J Hardy
- Centre for Phytophthora Science and Management, Murdoch University, Murdoch, WA, Australia
| | - Silke Jacques
- Centre for Crop and Disease Management, Curtin University, Bentley, WA, Australia
| | | | - Kar-Chun Tan
- Centre for Crop and Disease Management, Curtin University, Bentley, WA, Australia
| |
Collapse
|
17
|
Chang CF, Brown KM, Yang Y, Brugmann SA. Centriolar Protein C2cd3 Is Required for Craniofacial Development. Front Cell Dev Biol 2021; 9:647391. [PMID: 34211969 PMCID: PMC8239364 DOI: 10.3389/fcell.2021.647391] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2020] [Accepted: 05/17/2021] [Indexed: 11/13/2022] Open
Abstract
The primary cilium is a ubiquitous, microtubule-based cellular organelle. Primary cilia dysfunction results in a group of disorders termed ciliopathies. C2 domain containing 3 centriole elongation regulator (C2cd3), encodes a centriolar protein essential for ciliogenesis. Mutations in human C2CD3 are associated with the human ciliopathy Oral-Facial-Digital syndrome type 14 (OFD14). In order to better understand the etiology of ciliopathies including OFD14, we generated numerous murine models targeting C2cd3. Initial analysis revealed several tissue-specific isoforms of C2cd3, and while the loss of C2cd3 has previously been reported to result in exencephaly, tight mesencephalic flexure, pericardial edema, abnormal heart looping and a twisted body axis, further analysis revealed that genetic background may also contribute to phenotypic variation. Additional analyses of a conditional allelic series targeting C-terminal PKC-C2 domains or the N-terminal C2CD3N-C2 domain of C2cd3 revealed a variable degree of phenotypic severity, suggesting that while the N-terminal C2CD3N-C2 domain was critical for early embryonic development as a whole, there was also a craniofacial specific role for the C2CD3N-C2 domains. Together, through generation of novel models and evaluation of C2cd3 expression, these data provide valuable insight into mechanisms of pathology for craniofacial ciliopathies that can be further explored in the future.
Collapse
Affiliation(s)
- Ching-Fang Chang
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Kari M Brown
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Yanfen Yang
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States
| | - Samantha A Brugmann
- Division of Developmental Biology, Department of Pediatrics, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Department of Pediatrics, University of Cincinnati College of Medicine, Cincinnati, OH, United States.,Division of Plastic Surgery, Department of Surgery, Cincinnati Children's Hospital Medical Center, Cincinnati, OH, United States.,Shriners Children's Hospital, Cincinnati, OH, United States
| |
Collapse
|
18
|
Xu J, Ou X, Li J, Cai Q, Sun K, Ye J, Peng J. Overexpression of TC2N is associated with poor prognosis in gastric cancer. J Cancer 2021; 12:807-817. [PMID: 33403038 PMCID: PMC7778556 DOI: 10.7150/jca.50653] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2020] [Accepted: 10/30/2020] [Indexed: 12/27/2022] Open
Abstract
Background: Tac2-N (TC2N) is a tandem C2 domain-containing protein, acting as a novel oncogene or suppressor in different kinds of cancers. However, the status of TC2N expression and its significance in gastric cancer (GC) is still unclear. The present study is aimed to elucidate the clinicopathological significance and prognostic value of TC2N level in GC. Methods: We used sequencing data from the Cancer Genome Atlas (TCGA) database to analyze TC2N expression in GC by UALCAN database and Gene Expression Profiling Interactive Analysis tools (GEPIA). TC2N expression level in 12 pairs of fresh GC tissues and adjacent nontumorous tissues was detected by quantitative real-time reverse-transcription polymerase chain reaction (RT-PCR) and Western blot (WB) assays. Immunohistochemical (IHC) staining was used to detect TC2N protein expression in Paraffin-embedded tissues in our center. In vitro proliferation, migration and invasion assays were used to evaluate the effect of TC2N on functional capability of gastric cancer cells. LinkedOmics was used to identify gene expressions associated with TC2N. Results: The mRNA and protein expression of TC2N in gastric cancer were both significantly higher than normal gastric mucosa. It was also elevated in gastric cancer cells compared with normal gastric epithelium cell. In vitro assays suggested that TC2N facilitated proliferation, migration and invasion of gastric cancer cells. Bioinformatic analysis showed a widespread impact of TC2N on the transcriptome and a strong interaction with tumor associated genes. We also found that TC2N was an independent prognostic factor for long-term survival in GC patients and its high expression was evidently associated with poor overall survival and recurrence-free survival. Conclusions: Our results show that high level of TC2N correlates with poor prognosis in patients with gastric cancer and promotes the development of gastric cancer. Thus, TC2N expression can serve as a prognostic biomarker for patients with gastric cancer.
Collapse
Affiliation(s)
- Jianbo Xu
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
| | - Xinde Ou
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China.,Laboratory of General Surgery, the First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
| | - Jin Li
- Digestive Disease Center, the Seventh Affiliated Hospital, Sun Yat-sen University, 628 Zhenyuan Road, Shenzhen 518000, China.,Laboratory of General Surgery, the First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
| | - Qinbo Cai
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China.,Laboratory of General Surgery, the First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
| | - Kaiyu Sun
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
| | - Jingning Ye
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
| | - Jianjun Peng
- Department of Gastrointestinal Surgery, the First Affiliated Hospital, Sun Yat-sen University, 58 Zhongshan 2nd Road, Guangzhou 510080, China
| |
Collapse
|
19
|
Qureshi MA, Khan S, Tauheed MS, Ali Syed S, Ujjan ID, Lail A, Sharafat S. Pan-Cancer Multiomics Analysis of TC2N Gene Suggests its Important Role(s) in Tumourigenesis of Many Cancers. Asian Pac J Cancer Prev 2020; 21:3199-3209. [PMID: 33247676 PMCID: PMC8033114 DOI: 10.31557/apjcp.2020.21.11.3199] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2020] [Accepted: 11/22/2020] [Indexed: 12/20/2022] Open
Abstract
BACKGROUND Role of TC2N in carcinogenesis has been largely unfathomed until recently when it was identified as a novel oncogene in lung cancer. Subsequently, a tumour suppressor role of TC2N was reported in breast cancer. It is therefore highly relevant to investigate TC2N molecular partners/mechanisms on a larger scale including a wider range of tumour types. METHODS We investigated TC2N mRNA expression, its promoter methylation levels, effects of TC2N transcription on overall patient survival, somatic mutations in TC2N gene and correlation between TC2N mRNA expression and other cancer genes in pan-cancer by using data available from the Cancer Genome Atlas (TCGA) and the Genotype Tissue Expression (GTEx) databases. RESULTS TC2N mRNA expression was differentially regulated in 9/33 TCGA tumour types. Of these 9 tumours, 5 tumour types (cholangiocarcinoma, ovarian-serous-cystadenocarcinoma, rectal-adenocarcinoma, stomach-adenocarcinoma and thymoma) had significantly higher TC2N mRNA expression while 4 (pheochromocytoma-and-paraganglioma, skin-cutaneous-melanoma, thyroid-carcinoma and uterine-carcinosarcoma) had significantly lower TC2N mRNA expression compared to matched and normal controls. TC2N promoter was hypermethylated in most cancers while hypomethylated in head-and-neck-squamous-cell-carcinoma and kidney-renal-clear-cell carcinoma. TC2N transcription was positively correlated with transcription of several other cancer genes including genes from Myc, cell-cycle, Nrf2, Wnt, PI3K, Hippo, Notch, TGFβ and RAS/RTK pathways. Poor prognosis was associated with higher TC2N mRNA levels in pancreatic-adenocarcinoma and brain-lower-grade-glioma and lower TC2N mRNA levels in kidney-renal-clear-cell-carcinoma, mesothelioma, sarcoma and skin-cutaneous melanoma. Functional protein partners of TC2N were identified as STX2, SMEK1, SMEK2, STXBP5, SCARA5, MMRN1, CATSPER2, CATSPERB, CLEC4M and STAB2. Many of these proteins are key players in carcinogenesis of various cancers. Highest pathogenic somatic mutation rates in TC2N were found in skin-cutaneous-melanoma, uterine-corpus-endometrial-carcinoma, colon-endocervical-adenocarcinoma, bladder-urothelial-carcinoma and breast-invasive-carcinoma. CONCLUSION Our findings unravel several un-explored avenues related to the role of TC2N in tumourigenesis of several cancers, suggesting TC2N as an important player and a potential candidate for tumour-therapy.
Collapse
Affiliation(s)
- Muhammad Asif Qureshi
- Department of Pathology, Dow International Medical College, Dow University of Health Sciences Karachi, Pakistan.
| | - Saeed Khan
- Department of Pathology, Dow International Medical College, Dow University of Health Sciences Karachi, Pakistan.
| | - Muhammad Sohaib Tauheed
- Department of Pathology, Dow International Medical College, Dow University of Health Sciences Karachi, Pakistan.
| | - Sofia Ali Syed
- Department of Oral Pathology, Dow Dental College, Dow University of Health Sciences, Karachi, Pakistan.
| | - Ikram Din Ujjan
- Department of Pathology, Liaquat University of Medical and Health Sciences Jamshoro, Pakistan.
| | - Amanullah Lail
- Department of Paediatrics, Dow University of Health Sciences Karachi, Pakistan.
| | - Shaheen Sharafat
- Department of Pathology, Dow International Medical College, Dow University of Health Sciences Karachi, Pakistan.
| |
Collapse
|
20
|
Shen L, Zhang P, Wang J, Ji P. Tac2-N serves an oncogenic role and promotes drug resistance in human gastric cancer cells. Exp Ther Med 2020; 20:113. [PMID: 32989391 PMCID: PMC7517536 DOI: 10.3892/etm.2020.9241] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2020] [Accepted: 05/29/2020] [Indexed: 12/25/2022] Open
Abstract
Gastric cancer is one of the most common types of malignancy worldwide. Tac2-N (TC2N) has been reported to serve as either an oncogene or tumor suppressor in numerous different types of cancer; however, the role of TC2N in gastric cancer remains poorly understood. The present study aimed to investigate the role of TC2N in gastric cancer and reveal its regulatory mechanism. A Cell Counting Kit-8 assay was used to analyze the cell proliferation rate, while wound healing and Transwell Matrigel assays were performed to determine the cell migratory and invasive abilities, respectively. Cell cycle distribution was determined by flow cytometric analysis, and the expression levels of TC2N, P-glycoprotein (P-gp), cyclin D1, CDK4, cyclin E1, MMP2, MMP9 and N-Myc downstream regulated gene 1 were analyzed using reverse transcription-quantitative PCR or western blotting. Bioinformatics analysis revealed a high expression of TC2N in patients with gastric cancer. The experimental results revealed that TC2N expression levels were significantly unregulated in gastric cancer cell lines. The knockdown of TC2N in AGS cells significantly inhibited the cell proliferation rate and induced cell cycle arrest at the G0/G1 phase, while downregulating cyclin E1, cyclin D1 and CDK4 expression levels. The knockdown of TC2N also inhibited cell migration and invasion. Furthermore, the knockdown of TC2N improved the sensitivity of AGS cells to cisplatin, paclitaxel and 5-fluorouracil, and downregulated the protein expression levels of P-gp. By contrast, TC2N overexpression exerted the opposite effects in AGS cells. In conclusion, the findings of the present study indicated that the genetic knockdown of TC2N may inhibit cell proliferation, migration and invasion, while inducing cell cycle arrest in the G1/S phase and reversing the drug resistance of AGS cells, which may be partly through inhibiting P-gp expression levels. Thus, TC2N may serve as a novel diagnostic marker and therapeutic target for patients with gastric cancer.
Collapse
Affiliation(s)
- Lei Shen
- Department of Oncology, The Third People's Hospital of Hefei, Hefei, Anhui 230051, P.R. China
| | - Pingping Zhang
- Department of Physiology, Anhui Medical University, Hefei, Anhui 230032, P.R. China
| | - Jianbing Wang
- Department of Oncology, The Third People's Hospital of Hefei, Hefei, Anhui 230051, P.R. China
| | - Peng Ji
- Department of Magnetic Resonance, The Third People's Hospital of Hefei, Hefei, Anhui 230051, P.R. China
| |
Collapse
|
21
|
Quantitation and Simulation of Single Action Potential-Evoked Ca 2+ Signals in CA1 Pyramidal Neuron Presynaptic Terminals. eNeuro 2019; 6:ENEURO.0343-19.2019. [PMID: 31551250 PMCID: PMC6800293 DOI: 10.1523/eneuro.0343-19.2019] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2019] [Accepted: 09/10/2019] [Indexed: 01/07/2023] Open
Abstract
Presynaptic Ca2+ evokes exocytosis, endocytosis, and synaptic plasticity. However, Ca2+ flux and interactions at presynaptic molecular targets are difficult to quantify because fluorescence imaging has limited resolution. In rats of either sex, we measured single varicosity presynaptic Ca2+ using Ca2+ dyes as buffers, and constructed models of Ca2+ dispersal. Action potentials evoked Ca2+ transients with little variation when measured with low-affinity dye (peak amplitude 789 ± 39 nM, within 2 ms of stimulation; decay times, 119 ± 10 ms). Endogenous Ca2+ buffering capacity, action potential-evoked free [Ca2+]i, and total Ca2+ amounts entering terminals were determined using Ca2+ dyes as buffers. These data constrained Monte Carlo (MCell) simulations of Ca2+ entry, buffering, and removal. Simulations of experimentally-determined Ca2+ fluxes, buffered by simulated calbindin28K well fit data, and were consistent with clustered Ca2+ entry followed within 4 ms by diffusion throughout the varicosity. Repetitive stimulation caused free varicosity Ca2+ to sum. However, simulated in nanometer domains, its removal by pumps and buffering was negligible, while local diffusion dominated. Thus, Ca2+ within tens of nanometers of entry, did not accumulate. A model of synaptotagmin1 (syt1)-Ca2+ binding indicates that even with 10 µM free varicosity evoked Ca2+, syt1 must be within tens of nanometers of channels to ensure occupation of all its Ca2+-binding sites. Repetitive stimulation, evoking short-term synaptic enhancement, does not modify probabilities of Ca2+ fully occupying syt1’s C2 domains, suggesting that enhancement is not mediated by Ca2+-syt1 interactions. We conclude that at spatiotemporal scales of fusion machines, Ca2+ necessary for their activation is diffusion dominated.
Collapse
|
22
|
aPKC in neuronal differentiation, maturation and function. Neuronal Signal 2019; 3:NS20190019. [PMID: 32269838 PMCID: PMC7104321 DOI: 10.1042/ns20190019] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/04/2019] [Revised: 09/10/2019] [Accepted: 09/11/2019] [Indexed: 12/17/2022] Open
Abstract
The atypical Protein Kinase Cs (aPKCs)—PRKCI, PRKCZ and PKMζ—form a subfamily within the Protein Kinase C (PKC) family. These kinases are expressed in the nervous system, including during its development and in adulthood. One of the aPKCs, PKMζ, appears to be restricted to the nervous system. aPKCs are known to play a role in a variety of cellular responses such as proliferation, differentiation, polarity, migration, survival and key metabolic functions such as glucose uptake, that are critical for nervous system development and function. Therefore, these kinases have garnered a lot of interest in terms of their functional role in the nervous system. Here we review the expression and function of aPKCs in neural development and in neuronal maturation and function. Despite seemingly paradoxical findings with genetic deletion versus gene silencing approaches, we posit that aPKCs are likely candidates for regulating many important neurodevelopmental and neuronal functions, and may be associated with a number of human neuropsychiatric diseases.
Collapse
|
23
|
Hao X, Gao LY, Zhang N, Chen H, Jiang X, Liu W, Ao L, Cao J, Han F, Liu J. Tac2-N acts as a novel oncogene and promotes tumor metastasis via activation of NF-κB signaling in lung cancer. J Exp Clin Cancer Res 2019; 38:319. [PMID: 31466523 PMCID: PMC6716936 DOI: 10.1186/s13046-019-1316-7] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/26/2019] [Accepted: 07/08/2019] [Indexed: 12/18/2022] Open
Abstract
BACKGROUND High rates of recurrence and metastasis are the major cause of the poor outcomes for patients with lung cancer. In previous research, we have demonstrated that Tac2-N promotes tumor growth by suppressing p53 signaling in lung cancer. Beyond that, other biological functions and clinical significance of Tac2-N in lung cancer progression are still unknown. METHODS Tissue microarrays of 272 lung cancer patients were constructed to assess the association of Tac2-N expression and prognosis of lung cancer patients with different clinical stages. The protein expression of Tac2-N in metastatic and non-metastatic specimens were detected by IHC. In vitro migration and invasion and in vivo nude mice metastasis model were used to evaluate the effect of Tac2-N ectopic expression on metastasis capability of lung cancer cells. The downstream signaling pathway of Tac2-N was explored using luciferase reporter assays and WB. RESULTS The expression of Tac2-N was associated with advanced stages, but not with early stages (P = 0.513). Tac2-N expression is sharply overexpressed in metastatic tumors compared with non-metastatic tumors. In vitro and in vivo assays suggested that Tac2-N facilitated migration and invasion of lung cancer cells in vitro and promoted tumor metastasis in vivo. Mechanistically, Tac2-N increased the degradation of IκB by promoting its phosphorylation, and subsequently activated NF-κB activity by facilitating the nuclear translocation of NF-κB and stimulating the transcription of targets, MMP7 and MMP9. Notably, the C2B domain of Tac2-N was crucial for Tac2-N to activate NF-κB signal. Blockage of NF-κB by shRNA or inhibitor attenuates the function of Tac2-N in the promotion of metastasis. CONCLUSIONS Our study provided proof of principle to show that Tac2-N serves as a novel oncogene gene and plays an important role in the progression and metastasis of lung cancer.
Collapse
Affiliation(s)
- Xianglin Hao
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| | - Li-yun Gao
- School of Public Health, Xinxiang Medical University, Xinxiang, People’s Republic of China
- Cooperative innovation center of molecular diagnosis and medical inspection technology, Beijing, People’s Republic of China
| | - Ning Zhang
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| | - Hongqiang Chen
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| | - Xiao Jiang
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| | - Wenbin Liu
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| | - Lin Ao
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| | - Jia Cao
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| | - Fei Han
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| | - Jinyi Liu
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, 30 Gaotanyan Street, Shapingba District, Chongqing, 400038 People’s Republic of China
| |
Collapse
|
24
|
Jackson-Hayes L, Atiq Z, Betton B, Freyaldenhoven WT, Myers L, Olsen E, Hill TW. Aspergillus nidulans protein kinase C forms a complex with the formin SepA that is involved in apical growth and septation. Fungal Genet Biol 2018; 122:21-30. [PMID: 30391723 DOI: 10.1016/j.fgb.2018.10.002] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/16/2017] [Revised: 10/22/2018] [Accepted: 10/23/2018] [Indexed: 10/27/2022]
Abstract
The Aspergillus nidulans orthologue of Protein kinase C (PkcA) and the A. nidulans formin SepA participate in polarized growth. PkcA localizes to growing hyphal apices and septation sites, and amino acid sequences within PkcA that are required for PkcA to localize to these sites of cell wall synthesis have been identified. SepA is associated with the contractile actomyosin ring (CAR), and it localizes at hyphal tips in association with the Spitzenkörper (SPK) and as an apical dome. A mutation in the sepA gene (sepA1) renders A. nidulans aseptate at elevated temperature. Progress towards understanding the spatiotemporal relationship between PkcA and SepA during polarized growth is presented here. Fluorescent chimeras of PkcA and SepA strongly overlapped in some hyphal tips in a dome pattern, while other tips displayed SepA SPK and PkcA dome localization within the same tip. At septation sites PkcA and SepA consistently colocalized through late stages of CAR constriction. Bimolecular fluorescence complementation experimental results provide evidence that SepA and PkcA are both present in complexes at both hyphal tip domes and at cortical rings. A Gal4-based yeast two-hybrid analysis confirmed the physical interaction between SepA and PkcA, and indicted that the FH2 domain of SepA is involved in its physical interaction with PkcA. A functional interaction between PkcA and SepA was shown through complementation of the pkcA calC2 mutant's hypersensitivity to cell wall perturbing agents by overexpressed sepA and by the ability of the sepA1 mutation to block PkcA's ability to form cortical rings. Taken together these results suggest that a PkcA/SepA complex is involved in polarized growth. Through experiments using the actin disrupter latrunculin B, evidence is presented suggesting that actin plays a role in the PkcA/SepA complex.
Collapse
Affiliation(s)
- Loretta Jackson-Hayes
- Department of Chemistry, Rhodes College, 2000 N. Parkway, Memphis, TN 38112, USA; Biochemistry and Molecular Biology Program, Rhodes College, 2000 N. Parkway, Memphis, TN 38112, USA.
| | - Zainab Atiq
- Biochemistry and Molecular Biology Program, Rhodes College, 2000 N. Parkway, Memphis, TN 38112, USA
| | - Brianna Betton
- Biochemistry and Molecular Biology Program, Rhodes College, 2000 N. Parkway, Memphis, TN 38112, USA
| | - W Toler Freyaldenhoven
- Biochemistry and Molecular Biology Program, Rhodes College, 2000 N. Parkway, Memphis, TN 38112, USA
| | - Lance Myers
- Biochemistry and Molecular Biology Program, Rhodes College, 2000 N. Parkway, Memphis, TN 38112, USA
| | - Elisabet Olsen
- Department of Chemistry, Rhodes College, 2000 N. Parkway, Memphis, TN 38112, USA
| | - Terry W Hill
- Biochemistry and Molecular Biology Program, Rhodes College, 2000 N. Parkway, Memphis, TN 38112, USA; Department of Biology, Rhodes College, 2000 N. Parkway, Memphis, TN 38112, USA
| |
Collapse
|
25
|
Hao XL, Han F, Zhang N, Chen HQ, Jiang X, Yin L, Liu WB, Wang DD, Chen JP, Cui ZH, Ao L, Cao J, Liu JY. TC2N, a novel oncogene, accelerates tumor progression by suppressing p53 signaling pathway in lung cancer. Cell Death Differ 2018; 26:1235-1250. [PMID: 30254375 PMCID: PMC6748156 DOI: 10.1038/s41418-018-0202-8] [Citation(s) in RCA: 35] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2017] [Revised: 08/01/2018] [Accepted: 08/30/2018] [Indexed: 01/09/2023] Open
Abstract
The protein containing the C2 domain has been well documented for its essential roles in endocytosis, cellular metabolism and cancer. Tac2-N (TC2N) is a tandem C2 domain-containing protein, but its function, including its role in tumorigenesis, remains unknown. Here, we first identified TC2N as a novel oncogene in lung cancer. TC2N was preferentially upregulated in lung cancer tissues compared with adjacent normal lung tissues. High TC2N expression was significantly associated with poor outcome of lung cancer patients. Knockdown of TC2N markedly induces cell apoptosis and cell cycle arrest with repressing proliferation in vitro, and suppresses tumorigenicity in vivo, whereas overexpression of TC2N has the opposite effects both in vitro and in vivo. Using a combination of TCGA database and bioinformatics, we demonstrate that TC2N is involved in regulation of the p53 signaling pathway. Mechanistically, TC2N attenuates p53 signaling pathway through inhibiting Cdk5-induced phosphorylation of p53 via inducing Cdk5 degradation or disrupting the interaction between Cdk5 and p53. Moreover, the blockade of p53 attenuates the function of TC2N knockdown in the regulation of cell proliferation and apoptosis. In addition, downregulated TC2N is involved in the apoptosis of lung cancer cells induced by doxorubicin, leading to p53 pathway activation. Overall, these findings uncover a role for the p53 inactivator TC2N in regulating the proliferation and apoptosis of lung cancer cells. Our present study provides novel insights into the mechanism of tumorigenesis in lung cancer.
Collapse
Affiliation(s)
- Xiang-Lin Hao
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Fei Han
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Ning Zhang
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Hong-Qiang Chen
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Xiao Jiang
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Li Yin
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Wen-Bin Liu
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Dan-Dan Wang
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Jian-Ping Chen
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Zhi-Hong Cui
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Lin Ao
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China
| | - Jia Cao
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China.
| | - Jin-Yi Liu
- Institute of Toxicology, College of Preventive Medicine, Third Military Medical University, Chongqing, 400038, China.
| |
Collapse
|
26
|
Tsuchiya T, Fujii M, Matsuda N, Kunida K, Uda S, Kubota H, Konishi K, Kuroda S. System identification of signaling dependent gene expression with different time-scale data. PLoS Comput Biol 2017; 13:e1005913. [PMID: 29281625 PMCID: PMC5760096 DOI: 10.1371/journal.pcbi.1005913] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2017] [Revised: 01/09/2018] [Accepted: 12/01/2017] [Indexed: 01/11/2023] Open
Abstract
Cells decode information of signaling activation at a scale of tens of minutes by downstream gene expression with a scale of hours to days, leading to cell fate decisions such as cell differentiation. However, no system identification method with such different time scales exists. Here we used compressed sensing technology and developed a system identification method using data of different time scales by recovering signals of missing time points. We measured phosphorylation of ERK and CREB, immediate early gene expression products, and mRNAs of decoder genes for neurite elongation in PC12 cell differentiation and performed system identification, revealing the input-output relationships between signaling and gene expression with sensitivity such as graded or switch-like response and with time delay and gain, representing signal transfer efficiency. We predicted and validated the identified system using pharmacological perturbation. Thus, we provide a versatile method for system identification using data with different time scales.
Collapse
Affiliation(s)
- Takaho Tsuchiya
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Masashi Fujii
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
- Molecular Genetics Research Laboratory, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Naoki Matsuda
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
| | - Katsuyuki Kunida
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
- Laboratory of Computational Biology, Graduate School of Biological Sciences, Nara Institute of Science and Technology, Ikoma, Japan
| | - Shinsuke Uda
- Division of Integrated Omics, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Hiroyuki Kubota
- Division of Integrated Omics, Research Center for Transomics Medicine, Medical Institute of Bioregulation, Kyushu University, Fukuoka, Japan
| | - Katsumi Konishi
- Department of Computer Science, Faculty of Informatics, Kogakuin University, Tokyo, Japan
| | - Shinya Kuroda
- Department of Biological Sciences, Graduate School of Science, University of Tokyo, Tokyo, Japan
- CREST, Japan Science and Technology Corporation, Tokyo, Japan
| |
Collapse
|
27
|
Novel calpain families and novel mechanisms for calpain regulation in Aplysia. PLoS One 2017; 12:e0186646. [PMID: 29053733 PMCID: PMC5650170 DOI: 10.1371/journal.pone.0186646] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2017] [Accepted: 10/04/2017] [Indexed: 11/19/2022] Open
Abstract
Calpains are a family of intracellular proteases defined by a conserved protease domain. In the marine mollusk Aplysia californica, calpains are important for the induction of long-term synaptic plasticity and memory, at least in part by cleaving protein kinase Cs (PKCs) into constitutively active kinases, termed protein kinase Ms (PKMs). We identify 14 genes encoding calpains in Aplysia using bioinformatics, including at least one member of each of the four major calpain families into which metazoan calpains are generally classified, as well as additional truncated and atypical calpains. Six classical calpains containing a penta-EF-hand (PEF) domain are present in Aplysia. Phylogenetic analysis determined that these six calpains come from three separate classical calpain families. One of the classical calpains in Aplysia, AplCCal1, has been implicated in plasticity. We identify three splice cassettes and an alternative transcriptional start site in AplCCal1. We characterize several of the possible isoforms of AplCCal1 in vitro, and demonstrate that AplCCal1 can cleave PKCs into PKMs in a calcium-dependent manner in vitro. We also find that AplCCal1 has a novel mechanism of auto-inactivation through N-terminal cleavage that is modulated through its alternative transcriptional start site.
Collapse
|
28
|
Miyamoto A, Mikoshiba K. Probes for manipulating and monitoring IP 3. Cell Calcium 2016; 64:57-64. [PMID: 27887748 DOI: 10.1016/j.ceca.2016.11.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 11/14/2016] [Indexed: 12/31/2022]
Abstract
Inositol 1,4,5-trisphosphate (IP3) is an important second messenger produced via G-protein-coupled receptor- or receptor tyrosine kinase-mediated pathways. IP3 levels induce Ca2+ release from the endoplasmic reticulum (ER) via IP3 receptor (IP3R) located in the ER membrane. The resultant spatiotemporal pattern of Ca2+ signals regulates diverse cellular functions, including fertilization, gene expression, synaptic plasticity, and cell death. Therefore, monitoring and manipulating IP3 levels is important to elucidate not only the functions of IP3-mediated pathways but also the encoding mechanism of IP3R as a converter of intracellular signals from IP3 to Ca2+.
Collapse
Affiliation(s)
- Akitoshi Miyamoto
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan
| | - Katsuhiko Mikoshiba
- Laboratory for Developmental Neurobiology, RIKEN Brain Science Institute, 2-1 Hirosawa, Wako, Saitama 351-0198, Japan.
| |
Collapse
|
29
|
Kanwal A, Kasetti S, Putcha UK, Asthana S, Banerjee SK. Protein kinase C-mediated sodium glucose transporter 1 activation in precondition-induced cardioprotection. DRUG DESIGN DEVELOPMENT AND THERAPY 2016; 10:2929-2938. [PMID: 27695290 PMCID: PMC5028101 DOI: 10.2147/dddt.s105482] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
The concept of cardioprotection through preconditioning against ischemia-reperfusion (I/R) injury is well known and established. However, among different proposed mechanisms regarding the concept of ischemic preconditioning, protein kinase C (PKC)-mediated cardioprotection through ischemic preconditioning plays a key role in myocardial I/R injury. Thus, this study was designed to find the relationship between PKC and sodium glucose transporter 1 (SGLT1) in preconditioning-induced cardioprotection, which is ill reported till now. By applying a multifaceted approach, we demonstrated that PKC activates SGLT1, which curbed oxidative stress and apoptosis against I/R injury. PKC activation enhances cardiac glucose uptake through SGLT1 and seems essential in preventing I/R-induced cardiac injury, indicating a possible cross-talk between PKC and SGLT1.
Collapse
Affiliation(s)
- Abhinav Kanwal
- Division of Medicinal Chemistry and Pharmacology, Indian Institute of Chemical Technology, Hyderabad, India; Drug Discovery Research Center (DDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
| | - Sujatha Kasetti
- Department of Pharmacology, National Institute of Pharmaceutical Education and Research, Hyderabad, India
| | - Uday Kumar Putcha
- Department of Pathology, National Institute of Nutrition, Hyderabad, India
| | - Shailendra Asthana
- Drug Discovery Research Center (DDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
| | - Sanjay K Banerjee
- Division of Medicinal Chemistry and Pharmacology, Indian Institute of Chemical Technology, Hyderabad, India; Drug Discovery Research Center (DDRC), Translational Health Science and Technology Institute (THSTI), Faridabad, Haryana, India
| |
Collapse
|
30
|
Das J, Ramani R, Suraju MO. Polyphenol compounds and PKC signaling. Biochim Biophys Acta Gen Subj 2016; 1860:2107-21. [PMID: 27369735 DOI: 10.1016/j.bbagen.2016.06.022] [Citation(s) in RCA: 72] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2016] [Revised: 06/01/2016] [Accepted: 06/26/2016] [Indexed: 12/17/2022]
Abstract
BACKGROUND Naturally occurring polyphenols found in food sources provide huge health benefits. Several polyphenolic compounds are implicated in the prevention of disease states, such as cancer. One of the mechanisms by which polyphenols exert their biological actions is by interfering in the protein kinase C (PKC) signaling pathways. PKC belongs to a superfamily of serine-threonine kinase and are primarily involved in phosphorylation of target proteins controlling activation and inhibition of many cellular processes directly or indirectly. SCOPE OF REVIEW Despite the availability of substantial literature data on polyphenols' regulation of PKC, no comprehensive review article is currently available on this subject. This article reviews PKC-polyphenol interactions and its relevance to various disease states. In particular, salient features of polyphenols, PKC, interactions of naturally occurring polyphenols with PKC, and future perspective of research on this subject are discussed. MAJOR CONCLUSIONS Some polyphenols exert their antioxidant properties by regulating the transcription of the antioxidant enzyme genes through PKC signaling. Regulation of PKC by polyphenols is isoform dependent. The activation or inhibition of PKC by polyphenols has been found to be dependent on the presence of membrane, Ca(2+) ion, cofactors, cell and tissue types etc. Two polyphenols, curcumin and resveratrol are in clinical trials for the treatment of colon cancer. GENERAL SIGNIFICANCE The fact that 74% of the cancer drugs are derived from natural sources, naturally occurring polyphenols or its simple analogs with improved bioavailability may have the potential to be cancer drugs in the future.
Collapse
Affiliation(s)
- Joydip Das
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, United States.
| | - Rashmi Ramani
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, United States
| | - M Olufemi Suraju
- Department of Pharmacological and Pharmaceutical Sciences, College of Pharmacy, University of Houston, Houston, TX 77204, United States
| |
Collapse
|
31
|
Mittal R, Grati M, Yan D, Liu XZ. Pseudomonas aeruginosa Activates PKC-Alpha to Invade Middle Ear Epithelial Cells. Front Microbiol 2016; 7:255. [PMID: 26973629 PMCID: PMC4777741 DOI: 10.3389/fmicb.2016.00255] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2016] [Accepted: 02/16/2016] [Indexed: 12/31/2022] Open
Abstract
Otitis media (OM) is a group of complex inflammatory disorders affecting the middle ear which can be acute or chronic. Chronic suppurative otitis media (CSOM) is a form of chronic OM characterized by tympanic membrane perforation and discharge. Despite the significant impact of CSOM on human population, it is still an understudied and unexplored research area. CSOM is a leading cause of hearing loss and life-threatening central nervous system complications. Bacterial exposure especially Pseudomonas aeruginosa is the most common cause of CSOM. Our previous studies have demonstrated that P. aeruginosa invades human middle ear epithelial cells (HMEECs). However, molecular mechanisms leading to bacterial invasion of HMEECs are not known. The aim of this study is to characterize the role of PKC pathway in the ability of P. aeruginosa to colonize HMEECs. We observed that otopathogenic P. aeruginosa activates the PKC pathway, specifically phosphorylation of PKC-alpha (PKC-α) in HMEECs. The ability of otopathogenic P. aeruginosa to phosphorylate PKC-α depends on bacterial OprF expression. The activation of PKC-α was associated with actin condensation. Blocking the PKC pathway attenuated the ability of bacteria to invade HMEECs and subsequent actin condensation. This study, for the first time, demonstrates that the host PKC-α pathway is involved in invasion of HMEECs by P. aeruginosa and subsequently to cause OM. Characterizing the role of the host signaling pathway in the pathogenesis of CSOM will provide novel avenues to design effective treatment modalities against the disease.
Collapse
Affiliation(s)
- Rahul Mittal
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami Florida, USA
| | - M'hamed Grati
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami Florida, USA
| | - Denise Yan
- Department of Otolaryngology, University of Miami Miller School of Medicine, Miami Florida, USA
| | - Xue Z Liu
- Department of Otolaryngology, University of Miami Miller School of Medicine, MiamiFlorida, USA; Department of Biochemistry, University of Miami Miller School of Medicine, MiamiFL, USA; Department of Human Genetics, University of Miami Miller School of Medicine, MiamiFL, USA; Department of Otolaryngology, Xiangya Hospital, Central South UniversityChangsha, China
| |
Collapse
|
32
|
Abstract
KATP channels are integral to the functions of many cells and tissues. The use of electrophysiological methods has allowed for a detailed characterization of KATP channels in terms of their biophysical properties, nucleotide sensitivities, and modification by pharmacological compounds. However, even though they were first described almost 25 years ago (Noma 1983, Trube and Hescheler 1984), the physiological and pathophysiological roles of these channels, and their regulation by complex biological systems, are only now emerging for many tissues. Even in tissues where their roles have been best defined, there are still many unanswered questions. This review aims to summarize the properties, molecular composition, and pharmacology of KATP channels in various cardiovascular components (atria, specialized conduction system, ventricles, smooth muscle, endothelium, and mitochondria). We will summarize the lessons learned from available genetic mouse models and address the known roles of KATP channels in cardiovascular pathologies and how genetic variation in KATP channel genes contribute to human disease.
Collapse
Affiliation(s)
- Monique N Foster
- Departments of Pediatrics, Physiology & Neuroscience, and Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| | - William A Coetzee
- Departments of Pediatrics, Physiology & Neuroscience, and Biochemistry and Molecular Pharmacology, NYU School of Medicine, New York, New York
| |
Collapse
|
33
|
Lum MA, Barger CJ, Hsu AH, Leontieva OV, Black AR, Black JD. Protein Kinase Cα (PKCα) Is Resistant to Long Term Desensitization/Down-regulation by Prolonged Diacylglycerol Stimulation. J Biol Chem 2016; 291:6331-46. [PMID: 26769967 DOI: 10.1074/jbc.m115.696211] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/01/2015] [Indexed: 11/06/2022] Open
Abstract
Sustained activation of PKCα is required for long term physiological responses, such as growth arrest and differentiation. However, studies with pharmacological agonists (e.g. phorbol 12-myristate 13-acetate (PMA)) indicate that prolonged stimulation leads to PKCα desensitization via dephosphorylation and/or degradation. The current study analyzed effects of chronic stimulation with the physiological agonist diacylglycerol. Repeated addition of 1,2-dioctanoyl-sn-glycerol (DiC8) resulted in sustained plasma membrane association of PKCα in a pattern comparable with that induced by PMA. However, although PMA potently down-regulated PKCα, prolonged activation by DiC8 failed to engage known desensitization mechanisms, with the enzyme remaining membrane-associated and able to support sustained downstream signaling. DiC8-activated PKCα did not undergo dephosphorylation, ubiquitination, or internalization, early events in PKCα desensitization. Although DiC8 efficiently down-regulated novel PKCs PKCδ and PKCϵ, differences in Ca(2+) sensitivity and diacylglycerol affinity were excluded as mediators of the selective resistance of PKCα. Roles for Hsp/Hsc70 and Hsp90 were also excluded. PMA, but not DiC8, targeted PKCα to detergent-resistant membranes, and disruption of these domains with cholesterol-binding agents demonstrated a role for differential membrane compartmentalization in selective agonist-induced degradation. Chronic DiC8 treatment failed to desensitize PKCα in several cell types and did not affect PKCβI; thus, conventional PKCs appear generally insensitive to desensitization by sustained diacylglycerol stimulation. Consistent with this conclusion, prolonged (several-day) membrane association/activation of PKCα is seen in self-renewing epithelium of the intestine, cervix, and skin. PKCα deficiency affects gene expression, differentiation, and tumorigenesis in these tissues, highlighting the importance of mechanisms that protect PKCα from desensitization in vivo.
Collapse
Affiliation(s)
- Michelle A Lum
- From the Eppley Institute for Research in Cancer and Allied Diseases and the Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198-5950 and the Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York 14263
| | - Carter J Barger
- From the Eppley Institute for Research in Cancer and Allied Diseases and the Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198-5950 and
| | - Alice H Hsu
- From the Eppley Institute for Research in Cancer and Allied Diseases and the Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198-5950 and
| | - Olga V Leontieva
- the Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York 14263
| | - Adrian R Black
- From the Eppley Institute for Research in Cancer and Allied Diseases and the Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198-5950 and the Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York 14263
| | - Jennifer D Black
- From the Eppley Institute for Research in Cancer and Allied Diseases and the Fred and Pamela Buffet Cancer Center, University of Nebraska Medical Center, Omaha, Nebraska 68198-5950 and the Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, New York 14263
| |
Collapse
|
34
|
Thai TL, Yu L, Galarza-Paez L, Wu MM, Lam HYC, Bao HF, Duke BJ, Al-Khalili O, Ma HP, Liu B, Eaton DC. The Polarized Effect of Intracellular Calcium on the Renal Epithelial Sodium Channel Occurs as a Result of Subcellular Calcium Signaling Domains Maintained by Mitochondria. J Biol Chem 2015; 290:28805-11. [PMID: 26451045 DOI: 10.1074/jbc.m115.668293] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2015] [Indexed: 12/27/2022] Open
Abstract
The renal epithelial sodium channel (ENaC) provides regulated sodium transport in the distal nephron. The effects of intracellular calcium ([Ca(2+)]i) on this channel are only beginning to be elucidated. It appears from previous studies that the [Ca(2+)]i increases downstream of ATP administration may have a polarized effect on ENaC, where apical application of ATP and the subsequent [Ca(2+)]i increase have an inhibitory effect on the channel, whereas basolateral ATP and [Ca(2+)]i have a stimulatory effect. We asked whether this polarized effect of ATP is, in fact, reflective of a polarized effect of increased [Ca(2+)]i on ENaC and what underlying mechanism is responsible. We began by performing patch clamp experiments in which ENaC activity was measured during apical or basolateral application of ionomycin to increase [Ca(2+)]i near the apical or basolateral membrane, respectively. We found that ENaC does indeed respond to increased [Ca(2+)]i in a polarized fashion, with apical increases being inhibitory and basolateral increases stimulating channel activity. In other epithelial cell types, mitochondria sequester [Ca(2+)]i, creating [Ca(2+)]i signaling microdomains within the cell that are dependent on mitochondrial localization. We found that mitochondria localize in bands just beneath the apical and basolateral membranes in two different cortical collecting duct principal cell lines and in cortical collecting duct principal cells in mouse kidney tissue. We found that inhibiting mitochondrial [Ca(2+)]i uptake destroyed the polarized response of ENaC to [Ca(2+)]i. Overall, our data suggest that ENaC is regulated by [Ca(2+)]i in a polarized fashion and that this polarization is maintained by mitochondrial [Ca(2+)]i sequestration.
Collapse
Affiliation(s)
- Tiffany L Thai
- From the Department of Physiology, Emory University, Atlanta, Georgia 30322
| | - Ling Yu
- From the Department of Physiology, Emory University, Atlanta, Georgia 30322
| | - Laura Galarza-Paez
- From the Department of Physiology, Emory University, Atlanta, Georgia 30322
| | - Ming Ming Wu
- From the Department of Physiology, Emory University, Atlanta, Georgia 30322
| | - Ho Yin Colin Lam
- From the Department of Physiology, Emory University, Atlanta, Georgia 30322
| | - Hui Fang Bao
- From the Department of Physiology, Emory University, Atlanta, Georgia 30322
| | - Billie Jeanne Duke
- From the Department of Physiology, Emory University, Atlanta, Georgia 30322
| | - Otor Al-Khalili
- From the Department of Physiology, Emory University, Atlanta, Georgia 30322
| | - He-Ping Ma
- From the Department of Physiology, Emory University, Atlanta, Georgia 30322
| | - Bingchen Liu
- From the Department of Physiology, Emory University, Atlanta, Georgia 30322
| | - Douglas C Eaton
- From the Department of Physiology, Emory University, Atlanta, Georgia 30322
| |
Collapse
|
35
|
Plasma membrane and cytoskeleton dynamics during single-cell wound healing. BIOCHIMICA ET BIOPHYSICA ACTA-MOLECULAR CELL RESEARCH 2015. [DOI: 10.1016/j.bbamcr.2015.07.012] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
|
36
|
Lucke-Wold BP, Turner RC, Logsdon AF, Simpkins JW, Alkon DL, Smith KE, Chen YW, Tan Z, Huber JD, Rosen CL. Common mechanisms of Alzheimer's disease and ischemic stroke: the role of protein kinase C in the progression of age-related neurodegeneration. J Alzheimers Dis 2015; 43:711-24. [PMID: 25114088 PMCID: PMC4446718 DOI: 10.3233/jad-141422] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
Ischemic stroke and Alzheimer's disease (AD), despite being distinct disease entities, share numerous pathophysiological mechanisms such as those mediated by inflammation, immune exhaustion, and neurovascular unit compromise. An important shared mechanistic link is acute and chronic changes in protein kinase C (PKC) activity. PKC isoforms have widespread functions important for memory, blood-brain barrier maintenance, and injury repair that change as the body ages. Disease states accelerate PKC functional modifications. Mutated forms of PKC can contribute to neurodegeneration and cognitive decline. In some cases the PKC isoforms are still functional but are not successfully translocated to appropriate locations within the cell. The deficits in proper PKC translocation worsen stroke outcome and amyloid-β toxicity. Cross talk between the innate immune system and PKC pathways contribute to the vascular status within the aging brain. Unfortunately, comorbidities such as diabetes, obesity, and hypertension disrupt normal communication between the two systems. The focus of this review is to highlight what is known about PKC function, how isoforms of PKC change with age, and what additional alterations are consequences of stroke and AD. The goal is to highlight future therapeutic targets that can be applied to both the treatment and prevention of neurologic disease. Although the pathology of ischemic stroke and AD are different, the similarity in PKC responses warrants further investigation, especially as PKC-dependent events may serve as an important connection linking age-related brain injury.
Collapse
Affiliation(s)
- Brandon P. Lucke-Wold
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Ryan C. Turner
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Aric F. Logsdon
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, USA
| | - James W. Simpkins
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Daniel L. Alkon
- Blanchette Rockefeller Neurosciences Institute, Morgantown, WV, USA
| | - Kelly E. Smith
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, USA
| | - Yi-Wen Chen
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Zhenjun Tan
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
| | - Jason D. Huber
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Department of Basic Pharmaceutical Sciences, West Virginia University School of Pharmacy, Morgantown, WV, USA
| | - Charles L. Rosen
- Department of Neurosurgery, West Virginia University School of Medicine, Morgantown, WV, USA
- The Center for Neuroscience, West Virginia University School of Medicine, Morgantown, WV, USA
- Correspondence to: Charles L. Rosen, MD, PhD, Department of Neurosurgery, West Virginia University School of Medicine, One Medical Center Drive, Suite 4300, Health Sciences Center, PO Box 9183, Morgantown, WV 26506-9183, USA. Tel.: +1 304 293 5041; Fax: +1 304 293 4819;
| |
Collapse
|
37
|
Ann J, Yoon S, Baek J, Kim DH, Lewin NE, Hill CS, Blumberg PM, Lee J. Design and synthesis of protein kinase C epsilon selective diacylglycerol lactones (DAG-lactones). Eur J Med Chem 2014; 90:332-41. [PMID: 25437619 DOI: 10.1016/j.ejmech.2014.11.025] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/14/2014] [Revised: 10/29/2014] [Accepted: 11/11/2014] [Indexed: 10/24/2022]
Abstract
DAG-lactones afford a synthetically accessible, high affinity platform for probing structure activity relationships at the C1 regulatory domain of protein kinase C (PKC). Given the central role of PKC isoforms in cellular signaling, along with their differential biological activities, a critical objective is the design of isoform selective ligands. Here, we report the synthesis of a series of DAG-lactones varying in their side chains, with a particular focus on linoleic acid derivatives. We evaluated their selectivity for PKC epsilon versus PKC alpha both under standard lipid conditions (100% phosphatidylserine, PS) as well as in the presence of a nuclear membrane mimetic lipid mixture (NML). We find that selectivity for PKC epsilon versus PKC alpha tended to be enhanced in the presence of the nuclear membrane mimetic lipid mixture and, for our lead compound, report a selectivity of 32-fold.
Collapse
Affiliation(s)
- Jihyae Ann
- Laboratory of Medicinal Chemistry, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | - Suyoung Yoon
- Laboratory of Medicinal Chemistry, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | - Jisoo Baek
- Laboratory of Medicinal Chemistry, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | - Da Hye Kim
- Laboratory of Medicinal Chemistry, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea
| | - Nancy E Lewin
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Colin S Hill
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Peter M Blumberg
- Laboratory of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, MD 20892, USA
| | - Jeewoo Lee
- Laboratory of Medicinal Chemistry, Research Institute of Pharmaceutical Sciences, College of Pharmacy, Seoul National University, Seoul 151-742, Republic of Korea.
| |
Collapse
|
38
|
Affiliation(s)
- Joydip Das
- Department of Pharmacological
and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 521 Science and Research Building 2, Houston, Texas 77204, United States
| | - Ghazi M. Rahman
- Department of Pharmacological
and Pharmaceutical Sciences, College of Pharmacy, University of Houston, 521 Science and Research Building 2, Houston, Texas 77204, United States
| |
Collapse
|
39
|
Gjyshi O, Bottero V, Veettil MV, Dutta S, Singh VV, Chikoti L, Chandran B. Kaposi's sarcoma-associated herpesvirus induces Nrf2 during de novo infection of endothelial cells to create a microenvironment conducive to infection. PLoS Pathog 2014; 10:e1004460. [PMID: 25340789 PMCID: PMC4207826 DOI: 10.1371/journal.ppat.1004460] [Citation(s) in RCA: 65] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/19/2014] [Accepted: 09/10/2014] [Indexed: 12/30/2022] Open
Abstract
Kaposi's sarcoma-associated herpesvirus (KSHV) is the etiological agent of Kaposi's sarcoma (KS) and primary effusion B-cell lymphoma. KSHV induces reactive oxygen species (ROS) early during infection of human dermal microvascular endothelial (HMVEC-d) cells that are critical for virus entry. One of the downstream targets of ROS is nuclear factor E2-related factor 2 (Nrf2), a transcription factor with important anti-oxidative functions. Here, we show that KS skin lesions have high Nrf2 activity compared to healthy skin tissue. Within 30 minutes of de novo KSHV infection of HMVEC-d cells, we observed Nrf2 activation through ROS-mediated dissociation from its inhibitor Keap1, Ser-40 phosphorylation, and subsequent nuclear translocation. KSHV binding and consequent signaling through Src, PI3-K and PKC-ζ were also important for Nrf2 stability, phosphorylation and transcriptional activity. Although Nrf2 was dispensable for ROS homeostasis, it was essential for the induction of COX-2, VEGF-A, VEGF-D, Bcl-2, NQO1, GCS, HO1, TKT, TALDO and G6PD gene expression in KSHV-infected HMVEC-d cells. The COX-2 product PGE2 induced Nrf2 activity through paracrine and autocrine signaling, creating a feed-forward loop between COX-2 and Nrf2. vFLIP, a product of KSHV latent gene ORF71, induced Nrf2 and its target genes NQO1 and HO1. Activated Nrf2 colocalized with the KSHV genome as well as with the latency protein LANA-1. Nrf2 knockdown enhanced ORF73 expression while reducing ORF50 and other lytic gene expression without affecting KSHV entry or genome nuclear delivery. Collectively, these studies for the first time demonstrate that during de novo infection, KSHV induces Nrf2 through intricate mechanisms involving multiple signal molecules, which is important for its ability to manipulate host and viral genes, creating a microenvironment conducive to KSHV infection. Thus, Nrf2 is a potential attractive target to intervene in KSHV infection and the associated maladies. KSHV infection of endothelial cells in vivo causes Kaposi's sarcoma and understanding the steps involved in de novo KSHV infection of these cells and the consequences is important to develop therapies to counter KSHV pathogenesis. Infection of endothelial cells in vitro is preceded by the induction of a network of host signaling agents that are necessary for virus entry, gene expression and establishment of latency. Our previous studies have implicated reactive oxygen species (ROS) as part of this network. In the current study, we show that ROS activate Nrf2, a master transcriptional regulator of genes involved in ROS homeostasis, apoptosis, glucose metabolism and angiogenesis. Besides ROS, KSHV utilizes additional aspects of host signaling to induce Nrf2 activity. We also observed that infection of endothelial cells deficient in Nrf2 resulted in downregulation of multiple genes important in KSHV pathogenesis, such as COX-2 and VEGF, and affected proper expression of two hallmark KSHV genes, lytic ORF50 and latent ORF73. Taken together, this study is the first to demonstrate the importance of Nrf2 during de novo KSHV infection of endothelial cells, and establishes Nrf2 as an attractive therapeutic target to control KSHV infection, establishment of latency and the associated cancers.
Collapse
Affiliation(s)
- Olsi Gjyshi
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Virginie Bottero
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Mohanan Valliya Veettil
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Sujoy Dutta
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Vivek Vikram Singh
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Leela Chikoti
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
| | - Bala Chandran
- H. M. Bligh Cancer Research Laboratories, Department of Microbiology and Immunology, Chicago Medical School, Rosalind Franklin University of Medicine and Science, North Chicago, Illinois, United States of America
- * E-mail:
| |
Collapse
|
40
|
Knockdown of PKCε Expression Inhibits Growth, Induces Apoptosis and Decreases Invasiveness of Human Glioma Cells Partially Through Stat3. J Mol Neurosci 2014; 55:21-31. [DOI: 10.1007/s12031-014-0341-4] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2014] [Accepted: 05/22/2014] [Indexed: 12/21/2022]
|
41
|
Zhao Q, Zhang ZW, Liu CY, Xiao J, Li ZB, Wu SH, Wu YJ, Yang DS, Quan LP, Lei MS. Significance of expression of PKCα, Annexin A2 and S100A10 proteins in gastric cancer. Shijie Huaren Xiaohua Zazhi 2014; 22:1793-1800. [DOI: 10.11569/wcjd.v22.i13.1793] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [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 clinical significance of expression of protein kinase C (PKC), Annexin A2 and S100A10 proteins in gastric cancer and provide valuable data for finding diagnosis related proteins in gastric cancer.
METHODS: The expression of PKCα, Annexin A2 and S100A10 proteins was detected by Western blot in normal gastric mucosa and gastric cancer tissues. Moreover, their expression was analyzed by immunohistochemistry in a tissue array containing normal gastric mucosa and gastric cancer tissues. The clinicopathologic significance of their expression was evaluated.
RESULTS: Western blot analysis showed that the expression of PKCα, Annexin A2 and S100A10 proteins was significantly higher in gastric cancer tissue than in normal gastric mucosa tissue (P < 0.01 for all). Immunohistochemistry analysis showed that the positive expression rates of PKCα, Annexin A2 and S100A10 proteins were significantly lower in normal gastric mucosa tissue than in gastric cancer tissue [8.82% (3/34) vs 76.54% (62/81), 5.88% (2/34) vs 79.01% (64/81), 2.94% (1/34) vs 59.26% (48/81); P < 0.01 for all].
CONCLUSION: The expression of PKCα, Annexin A2 and S100A10 proteins is up-regulated in gastric cancer tissue compared with normal gastric mucosa tissue and may be related with the occurrence and differentiation degree of gastric cancer.
Collapse
|
42
|
Assessing the subcellular distribution of oncogenic phosphoinositide 3-kinase using microinjection into live cells. Biosci Rep 2014; 34:BSR20130133. [PMID: 27919038 PMCID: PMC3985441 DOI: 10.1042/bsr20130133] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2013] [Revised: 02/18/2014] [Accepted: 03/04/2014] [Indexed: 02/07/2023] Open
Abstract
Oncogenic mutations in PIK3CA lead to an increase in intrinsic phosphoinositide kinase activity, but it is thought that increased access of PI3Kα (phosphoinositide 3-kinase α) to its PM (plasma membrane) localized substrate is also required for increased levels of downstream PIP3/Akt [phosphoinositide-3,4,5-trisphosphate/also called PKB (protein kinase B)] signalling. We have studied the subcellular localization of wild-type and the two most common oncogenic mutants of PI3Kα in cells maintained in growth media, and starved or stimulated cells using a novel method in which PI3Kα is pre-formed as a 1:1 p110α:p85α complex in vitro then introduced into live cells by microinjection. Oncogenic E545K and H1047R mutants did not constitutively interact with membrane lipids in vitro or in cells maintained in 10% (v/v) FBS. Following stimulation of RTKs (receptor tyrosine kinases), microinjected PI3Kα was recruited to the PM, but oncogenic forms of PI3Kα were not recruited to the PM to a greater extent and did not reside at the PM longer than the wild-type PI3Kα. Instead, the E545K mutant specifically bound activated Cdc42 in vitro and microinjection of E545K was associated with the formation of cellular protrusions, providing some preliminary evidence that changes in protein–protein interactions may play a role in the oncogenicity of the E545K mutant in addition to the well-known changes in lipid kinase activity. Oncogenic forms of PI3Kα (phosphoinositide 3-kinase α) microinjected into live cells are not recruited to the PM (plasma membrane) to a greater extent, and do not reside at the PM longer, than wild-type PI3Kα.
Collapse
|
43
|
Classical protein kinases C are regulated by concerted interaction with lipids: the importance of phosphatidylinositol-4,5-bisphosphate. Biophys Rev 2013; 6:3-14. [PMID: 28509956 DOI: 10.1007/s12551-013-0125-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2013] [Accepted: 10/03/2013] [Indexed: 10/26/2022] Open
Abstract
Classical protein kinase C (PKC) enzymes are known to be important factors in cell physiology both in terms of health and disease. They are activated by triggering signals that induce their translocation to membranes. The consensus view is that several secondary messengers are involved in this activation, such as cytosolic Ca2+ and diacylglycerol. Cytosolic Ca2+ bridges the C2 domain to anionic phospholipids as phosphatidylserine in the membrane, and diacylglycerol binds to the C1 domain. Both diacylglycerol and the increase in Ca2+ concentration are assumed to arise from the extracellular signal that triggers the hydrolysis of phosphatidylinositol-4,5-bisphosphate. However, results obtained during the last decade indicate that this phosphoinositide itself is also responsible for modulating classical PKC activity and its localization in the plasma membrane.
Collapse
|
44
|
Uda S, Saito TH, Kudo T, Kokaji T, Tsuchiya T, Kubota H, Komori Y, Ozaki YI, Kuroda S. Robustness and compensation of information transmission of signaling pathways. Science 2013; 341:558-61. [PMID: 23908238 DOI: 10.1126/science.1234511] [Citation(s) in RCA: 93] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Robust transmission of information despite the presence of variation is a fundamental problem in cellular functions. However, the capability and characteristics of information transmission in signaling pathways remain poorly understood. We describe robustness and compensation of information transmission of signaling pathways at the cell population level. We calculated the mutual information transmitted through signaling pathways for the growth factor-mediated gene expression. Growth factors appeared to carry only information sufficient for a binary decision. Information transmission was generally more robust than average signal intensity despite pharmacological perturbations, and compensation of information transmission occurred. Information transmission to the biological output of neurite extension appeared robust. Cells may use information entropy as information so that messages can be robustly transmitted despite variation in molecular activities among individual cells.
Collapse
Affiliation(s)
- Shinsuke Uda
- Department of Biophysics and Biochemistry, Graduate School of Science, University of Tokyo, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
A novel predicted calcium-regulated kinase family implicated in neurological disorders. PLoS One 2013; 8:e66427. [PMID: 23840464 PMCID: PMC3696010 DOI: 10.1371/journal.pone.0066427] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2012] [Accepted: 05/08/2013] [Indexed: 12/03/2022] Open
Abstract
The catalogues of protein kinases, the essential effectors of cellular signaling, have been charted in Metazoan genomes for a decade now. Yet, surprisingly, using bioinformatics tools, we predicted protein kinase structure for proteins coded by five related human genes and their Metazoan homologues, the FAM69 family. Analysis of three-dimensional structure models and conservation of the classic catalytic motifs of protein kinases present in four out of five human FAM69 proteins suggests they might have retained catalytic phosphotransferase activity. An EF-hand Ca2+-binding domain in FAM69A and FAM69B proteins, inserted within the structure of the kinase domain, suggests they may function as Ca2+-dependent kinases. The FAM69 genes, FAM69A, FAM69B, FAM69C, C3ORF58 (DIA1) and CXORF36 (DIA1R), are by large uncharacterised molecularly, yet linked to several neurological disorders in genetics studies. The C3ORF58 gene is found deleted in autism, and resides in the Golgi. Unusually high cysteine content and presence of signal peptides in some of the family members suggest that FAM69 proteins may be involved in phosphorylation of proteins in the secretory pathway and/or of extracellular proteins.
Collapse
|
46
|
Schwingshackl A, Teng B, Ghosh M, Lim KG, Tigyi G, Narayanan D, Jaggar JH, Waters CM. Regulation of interleukin-6 secretion by the two-pore-domain potassium channel Trek-1 in alveolar epithelial cells. Am J Physiol Lung Cell Mol Physiol 2012; 304:L276-86. [PMID: 23275623 DOI: 10.1152/ajplung.00299.2012] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
We recently proposed a role for the two-pore-domain K(+) (K2P) channel Trek-1 in the regulation of cytokine release from mouse alveolar epithelial cells (AECs) by demonstrating decreased interleukin-6 (IL-6) secretion from Trek-1-deficient cells, but the underlying mechanisms remained unknown. This study was designed to investigate the mechanisms by which Trek-1 decreases IL-6 secretion. We hypothesized that Trek-1 regulates tumor necrosis factor-α (TNF-α)-induced IL-6 release via NF-κB-, p38-, and PKC-dependent pathways. We found that Trek-1 deficiency decreased IL-6 secretion from mouse and human AECs at both transcriptional and translational levels. While NF-κB/p65 phosphorylation was unchanged, p38 phosphorylation was decreased in Trek-1-deficient cells, and pharmacological inhibition of p38 decreased IL-6 secretion in control but not Trek-1-deficient cells. Similarly, pharmacological inhibition of PKC also decreased IL-6 release, and we found decreased phosphorylation of the isoforms PKC/PKDμ (Ser(744/748)), PKCθ, PKCδ, PKCα/βII, and PKCζ/λ, but not PKC/PKDμ (Ser(916)) in Trek-1-deficient AECs. Phosphorylation of PKCθ, a Ca(2+)-independent isoform, was intact in control cells but impaired in Trek-1-deficient cells. Furthermore, TNF-α did not elevate the intracellular Ca(2+) concentration in control or Trek-1-deficient cells, and removal of extracellular Ca(2+) did not impair IL-6 release. In summary, we report the expression of Trek-1 in human AECs and propose that Trek-1 deficiency may alter both IL-6 translation and transcription in AECs without affecting Ca(2+) signaling. The results of this study identify Trek-1 as a new potential target for the development of novel treatment strategies against acute lung injury.
Collapse
Affiliation(s)
- Andreas Schwingshackl
- Department of Pediatrics, University of Tennessee Health Science Center, Memphis, TN 38103, USA.
| | | | | | | | | | | | | | | |
Collapse
|