1
|
Tyagi K, Roy A, Mandal S. Protein kinase C iota promotes glycolysis via PI3K/AKT/mTOR signalling in high grade serous ovarian cancer. Mol Biol Rep 2024; 51:983. [PMID: 39276277 DOI: 10.1007/s11033-024-09918-3] [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: 06/19/2024] [Accepted: 09/05/2024] [Indexed: 09/16/2024]
Abstract
BACKGROUND Epithelial ovarian cancer, especially high grade serous ovarian cancer (HGSOC) is by far, the most lethal gynecological malignancy with poor prognosis and high relapse rate. Despite of availability of several therapeutic interventions including poly-ADP ribose polymerase (PARP) inhibitors, HGSOC remains unmanageable and identification of early detection biomarkers and therapeutic targets for this lethal malady is highly warranted. Aberrant expression of protein kinase C iota (PKCί) is implicated in many cellular and physiological functions involved in tumorigenesis including cell proliferation and cell cycle deregulation. METHODS AND RESULTS Two high grade serous ovarian cancer cells SKOV3 and COV362 were employed in this study. PKCί was genetically knocked down or pharmacologically inhibited and several functional and biochemical assays were performed. We report that PKCί is overexpressed in HGSOC cells and patient tissue samples with a significant prognostic value. Pharmacological inhibition of PKCί by Na-aurothiomalate or its shRNA-mediated genetic knockdown suppressed HGSOC cell proliferation, EMT and induced apoptosis. Moreover, PKCί positively regulated GLUT1 and several other glycolytic genes including HK1, HK2, PGK1, ENO1 and LDHA to promote elevated glucose uptake and glycolysis in HGSOC cells. Mechanistically, PKCί drove glycolysis via PI3K/AKT/mTOR signalling. Na-aurothiomalate and highly selective, dual PI3K/mTOR inhibitor dactolisib could serve as novel anti-glycolytic drugs in HGSOC. CONCLUSION Taken together, our results indicate PKCί/PI3K/AKT/mTOR signalling cascade could be a novel therapeutic target in a lethal pathology like HGSOC.
Collapse
Affiliation(s)
- Komal Tyagi
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India
| | - Adhiraj Roy
- Amity Institute of Molecular Medicine & Stem Cell Research, Amity University, Sector 125, Noida, Uttar Pradesh, 201303, India.
| | - Supratim Mandal
- Department of Microbiology, University of Kalyani, Kalyani, Nadia, West Bengal, 741235, India
| |
Collapse
|
2
|
He Y, Qu L. Non-coding RNAs in diabetic peripheral neuropathy: their role and mechanisms underlying their effects. Metabolism 2024; 154:155833. [PMID: 38462040 DOI: 10.1016/j.metabol.2024.155833] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/28/2023] [Revised: 02/20/2024] [Accepted: 03/05/2024] [Indexed: 03/12/2024]
Abstract
Diabetic peripheral neuropathy (DPN) is a complication of diabetes with a high rate of disability. However, current clinical treatments for DPN are suboptimal. Non-coding RNAs (ncRNAs) are a type of RNAs that are not translated into proteins. NcRNAs perform functions that regulate epigenetic modifications, transcriptional or post-transcriptional regulators of proteins, and thus participate in the physiological and pathological processes of the body. NcRNAs play a role in the progress of DPN by affecting the processes of inflammation, oxidative stress, cellular autophagy or apoptosis. Therefore, ncRNAs treatment is regarded as a promising therapeutic approach for DPN. In addition, since some ncRNAs present stably in the blood of DPN patients, they are considered as potential biomarkers that contribute to early clinical diagnosis. In this paper, we review the studies on the role of ncRNAs in DPN in the last decade, and discuss the mechanisms of ncRNAs, aiming to provide a reference for the future research on the treatment and early diagnosis of DPN.
Collapse
Affiliation(s)
- Yiqian He
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Science, 100730 Beijing, China
| | - Ling Qu
- Department of Traditional Chinese Medicine, Peking Union Medical College Hospital, Peking Union Medical College, Chinese Academy of Medical Science, 100730 Beijing, China.
| |
Collapse
|
3
|
Kazanietz MG, Cooke M. Protein kinase C signaling "in" and "to" the nucleus: Master kinases in transcriptional regulation. J Biol Chem 2024; 300:105692. [PMID: 38301892 PMCID: PMC10907189 DOI: 10.1016/j.jbc.2024.105692] [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: 10/23/2023] [Revised: 01/08/2024] [Accepted: 01/09/2024] [Indexed: 02/03/2024] Open
Abstract
PKC is a multifunctional family of Ser-Thr kinases widely implicated in the regulation of fundamental cellular functions, including proliferation, polarity, motility, and differentiation. Notwithstanding their primary cytoplasmic localization and stringent activation by cell surface receptors, PKC isozymes impel prominent nuclear signaling ultimately impacting gene expression. While transcriptional regulation may be wielded by nuclear PKCs, it most often relies on cytoplasmic phosphorylation events that result in nuclear shuttling of PKC downstream effectors, including transcription factors. As expected from the unique coupling of PKC isozymes to signaling effector pathways, glaring disparities in gene activation/repression are observed upon targeting individual PKC family members. Notably, specific PKCs control the expression and activation of transcription factors implicated in cell cycle/mitogenesis, epithelial-to-mesenchymal transition and immune function. Additionally, PKCs isozymes tightly regulate transcription factors involved in stepwise differentiation of pluripotent stem cells toward specific epithelial, mesenchymal, and hematopoietic cell lineages. Aberrant PKC expression and/or activation in pathological conditions, such as in cancer, leads to profound alterations in gene expression, leading to an extensive rewiring of transcriptional networks associated with mitogenesis, invasiveness, stemness, and tumor microenvironment dysregulation. In this review, we outline the current understanding of PKC signaling "in" and "to" the nucleus, with significant focus on established paradigms of PKC-mediated transcriptional control. Dissecting these complexities would allow the identification of relevant molecular targets implicated in a wide spectrum of diseases.
Collapse
Affiliation(s)
- Marcelo G Kazanietz
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| | - Mariana Cooke
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania, USA.
| |
Collapse
|
4
|
Samanta P, Bhowmik A, Biswas S, Sarkar R, Ghosh R, Pakhira S, Mondal M, Sen S, Saha P, Hajra S. Therapeutic Effectiveness of Anticancer Agents Targeting Different Signaling Molecules Involved in Asymmetric Division of Cancer Stem Cell. Stem Cell Rev Rep 2023:10.1007/s12015-023-10523-3. [PMID: 36952080 DOI: 10.1007/s12015-023-10523-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/25/2023] [Indexed: 03/24/2023]
Abstract
Intra-tumoral heterogeneity is maintained by cancer stem cells (CSCs) with dysregulated self-renewal and asymmetric cell division (ACD). According to the cancer stem cell theory, by ACD a CSC can generate two daughter progenies with different fates such as one cancer stem cell and one differentiated cell. Therefore, this type of mitotic division supports vital process of the maintenance of CSC population. But this CSC pool reservation by ACD complicates the treatment of cancer patients, as CSCs give rise to aggressive clones which are prone to metastasis and drug-insensitivity. Hence, identification of therapeutic modalities which can target ACD of cancer stem cell is an intriguing part of cancer research. In this review, other than the discussion about the extrinsic inducers of ACD role of different proteins, miRNAs and lncRNAs in this type of cell division is also mentioned. Other than these, mode of action of the proven and potential drugs targeting ACD of CSC is also discussed here.
Collapse
Affiliation(s)
- Priya Samanta
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Arijit Bhowmik
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India.
| | - Souradeep Biswas
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Rupali Sarkar
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Rituparna Ghosh
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Shampa Pakhira
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Mrinmoyee Mondal
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Soummadeep Sen
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Prosenjit Saha
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India
| | - Subhadip Hajra
- Department of Cancer Chemoprevention, Chittaranjan National Cancer Institute (CNCI), 37, Shyama Prasad Mukherjee Rd, Bakul Bagan, Bhowanipore, Kolkata, West Bengal, 700026, India.
| |
Collapse
|
5
|
Investigation of UTR Variants by Computational Approaches Reveal Their Functional Significance in PRKCI Gene Regulation. Genes (Basel) 2023; 14:genes14020247. [PMID: 36833174 PMCID: PMC9956319 DOI: 10.3390/genes14020247] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2022] [Revised: 01/02/2023] [Accepted: 01/16/2023] [Indexed: 01/20/2023] Open
Abstract
Single nucleotide polymorphisms (SNPs) are associated with many diseases including neurological disorders, heart diseases, diabetes, and different types of cancers. In the context of cancer, the variations within non-coding regions, including UTRs, have gained utmost importance. In gene expression, translational regulation is as important as transcriptional regulation for the normal functioning of cells; modification in normal functions can be associated with the pathophysiology of many diseases. UTR-localized SNPs in the PRKCI gene were evaluated using the PolymiRTS, miRNASNP, and MicroSNIper for association with miRNAs. Furthermore, the SNPs were subjected to analysis using GTEx, RNAfold, and PROMO. The genetic intolerance to functional variation was checked through GeneCards. Out of 713 SNPs, a total of thirty-one UTR SNPs (three in 3' UTR region and twenty-nine in 5' UTR region) were marked as ≤2b by RegulomeDB. The associations of 23 SNPs with miRNAs were found. Two SNPs, rs140672226 and rs2650220, were significantly linked with expression in the stomach and esophagus mucosa. The 3' UTR SNPs rs1447651774 and rs115170199 and the 5' UTR region variants rs778557075, rs968409340, and 750297755 were predicted to destabilize the mRNA structure with substantial change in free energy (∆G). Seventeen variants were predicted to have linkage disequilibrium with various diseases. The SNP rs542458816 in 5' UTR was predicted to put maximum influence on transcription factor binding sites. Gene damage index(GDI) and loss of function (o:e) ratio values for PRKCI suggested that the gene is not tolerant to loss of function variants. Our results highlight the effects of 3' and 5' UTR SNP on miRNA, transcription and translation of PRKCI. These analyses suggest that these SNPs can have substantial functional importance in the PRKCI gene. Future experimental validation could provide further basis for the diagnosis and therapeutics of various diseases.
Collapse
|
6
|
Abdelatty A, Sun Q, Hu J, Wu F, Wei G, Xu H, Zhou G, Wang X, Xia H, Lan L. Pan-Cancer Study on Protein Kinase C Family as a Potential Biomarker for the Tumors Immune Landscape and the Response to Immunotherapy. Front Cell Dev Biol 2022; 9:798319. [PMID: 35174160 PMCID: PMC8841516 DOI: 10.3389/fcell.2021.798319] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2021] [Accepted: 11/24/2021] [Indexed: 11/27/2022] Open
Abstract
The protein kinase C (PKC) family has been described with its role in some cancers, either as a promoter or suppressor. PKC signaling also regulates a molecular switch between transactivation and transrepression activity of the peroxisome proliferator-activated receptor alpha (PPARalpha). However, the role of different PKC enzymes in tumor immunity remains poorly defined. This study aims to investigate the correlation between PKC genes and tumor immunity, in addition to studying the probability of their use as predictive biomarkers for tumor immunity and immunotherapeutic response. The ssGSEA and the ESTIMATE methods were used to assess 28 tumor-infiltrating lymphocytes (TILs) and the immune component of each cancer, then correlated with PKC levels. Prediction of PKC levels-dependent immunotherapeutic response was based on human leukocytic antigen (HLA) gene enrichment scores and programmed cell death 1 ligand (PD-L1) expression. Univariate and multivariate Cox analysis was performed to evaluate the prognostic role of PKC genes in cancers. Methylation level and CNAs could drive the expression levels of some PKC members, especially PRKCI, whose CNGs are predicted to elevate their level in many cancer types. The most crucial finding in this study was that PKC isoenzymes are robust biomarkers for the tumor immune status, PRKCB, PRKCH, and PRKCQ as stimulators, while PRKCI and PRKCZ as inhibitors in most cancers. Also, PKC family gene levels can be used as predictors for the response to immunotherapies, especially HLAs dependent and PD-L1 blockade-dependent ones. In addition to its prognostic function, all PKC family enzymes are promising tumor immunity biomarkers and can help select suitable immune therapy in different cancers.
Collapse
Affiliation(s)
- Alaa Abdelatty
- Department of Pathology in the School of Basic Medical Sciences and Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, China
- Department of Pathology, Faculty of Veterinary Medicine, Kafrelsheikh University, Kafrelsheikh, Egypt
| | - Qi Sun
- Department of Pathology, Nanjing Drum Tower Hospital, The Affiliated Hospital of Nanjing University Medical School, Nanjing, China
| | - Junhong Hu
- Department of Colorectal and Anal Surgery, The First Affiliated Hospital of Zhengzhou University, Zhengzhou, China
| | - Fubing Wu
- Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
| | - Guanqun Wei
- Department of Pathology in the School of Basic Medical Sciences and Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, China
| | - Haojun Xu
- Department of Pathology in the School of Basic Medical Sciences and Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, China
| | - Guoren Zhou
- Department of Oncology, Jiangsu Cancer Hospital and The Affiliated Cancer Hospital of Nanjing Medical University and Jiangsu Institute of Cancer Research, Nanjing, China
- *Correspondence: Guoren Zhou, ; Xiaoming Wang, ; Hongping Xia, ; Linhua Lan,
| | - Xiaoming Wang
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- *Correspondence: Guoren Zhou, ; Xiaoming Wang, ; Hongping Xia, ; Linhua Lan,
| | - Hongping Xia
- Department of Pathology in the School of Basic Medical Sciences and Key Laboratory of Antibody Technique of National Health Commission, Nanjing Medical University, Nanjing, China
- Sir Run Run Hospital, Nanjing Medical University, Nanjing, China
- Department of Hepatobiliary Surgery, The First Affiliated Hospital of Wannan Medical College (Yijishan Hospital of Wannan Medical College), Wuhu, China
- *Correspondence: Guoren Zhou, ; Xiaoming Wang, ; Hongping Xia, ; Linhua Lan,
| | - Linhua Lan
- Key Laboratory of Diagnosis and Treatment of Severe Hepato-Pancreatic Diseases of Zhejiang Province, The First Affiliated Hospital of Wenzhou Medical University, Wenzhou, China
- *Correspondence: Guoren Zhou, ; Xiaoming Wang, ; Hongping Xia, ; Linhua Lan,
| |
Collapse
|
7
|
van der Westhuizen D, Bezuidenhout DI, Munro OQ. Cancer molecular biology and strategies for the design of cytotoxic gold(I) and gold(III) complexes: a tutorial review. Dalton Trans 2021; 50:17413-17437. [PMID: 34693422 DOI: 10.1039/d1dt02783b] [Citation(s) in RCA: 6] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
This tutorial review highlights key principles underpinning the design of selected metallodrugs to target specific biological macromolecules (DNA and proteins). The review commences with a descriptive overview of the eukaryotic cell cycle and the molecular biology of cancer, particularly apoptosis, which is provided as a necessary foundation for the discovery, design, and targeting of metal-based anticancer agents. Drugs which target DNA have been highlighted and clinically approved metallodrugs discussed. A brief history of the development of mainly gold-based metallodrugs is presented prior to addressing ligand systems for stabilizing and adding functionality to bio-active gold(I) and gold(III) complexes, particularly in the burgeoning field of anticancer metallodrugs. Concepts such as multi-modal and selective cytotoxic agents are covered where necessary for selected compounds. The emerging role of carbenes as the ligand system of choice to achieve these goals for gold-based metallodrug candidates is highlighted prior to closing the review with comments on some future directions that this research field might follow. The latter section ultimately emphasizes the importance of understanding the fate of metal complexes in cells to garner key mechanistic insights.
Collapse
Affiliation(s)
- Danielle van der Westhuizen
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, Johannesburg 2050, South Africa.
| | - Daniela I Bezuidenhout
- Laboratory of Inorganic Chemistry, Environmental and Chemical Engineering, University of Oulu, P. O. Box 3000, 90014 Oulu, Finland.
| | - Orde Q Munro
- Molecular Sciences Institute, School of Chemistry, University of the Witwatersrand, Johannesburg 2050, South Africa.
| |
Collapse
|
8
|
Kim HK, Vasileva EA, Mishchenko NP, Fedoreyev SA, Han J. Multifaceted Clinical Effects of Echinochrome. Mar Drugs 2021; 19:412. [PMID: 34436251 PMCID: PMC8400489 DOI: 10.3390/md19080412] [Citation(s) in RCA: 27] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2021] [Revised: 07/21/2021] [Accepted: 07/23/2021] [Indexed: 01/21/2023] Open
Abstract
The marine drug histochrome is a special natural antioxidant. The active substance of the drug is echinochrome A (Ech A, 7-ethyl-2,3,5,6,8-pentahydroxy-1,4-naphthoquinone), the most abundant quinonoid pigment in sea urchins. The medicine is clinically used in cardiology and ophthalmology based on the unique properties of Ech A, which simultaneously block various links of free radical reactions. In the last decade, numerous studies have demonstrated the effectiveness of histochrome in various disease models without adverse effects. Here, we review the data on the various clinical effects and modes of action of Ech A in ophthalmic, cardiovascular, cerebrovascular, inflammatory, metabolic, and malignant diseases.
Collapse
Affiliation(s)
- Hyoung Kyu Kim
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutic Center, Department of Physiology, College of Medicine, Inje University, Busan 57392, Korea;
- Department of Health Sciences and Technology, Graduate School of Inje University, Busan 57392, Korea
| | - Elena A. Vasileva
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Prospect 100 let Vladivostoku, 159, 690022 Vladivostok, Russia; (E.A.V.); (N.P.M.); (S.A.F.)
| | - Natalia P. Mishchenko
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Prospect 100 let Vladivostoku, 159, 690022 Vladivostok, Russia; (E.A.V.); (N.P.M.); (S.A.F.)
| | - Sergey A. Fedoreyev
- G.B. Elyakov Pacific Institute of Bioorganic Chemistry, Far-Eastern Branch of the Russian Academy of Science, Prospect 100 let Vladivostoku, 159, 690022 Vladivostok, Russia; (E.A.V.); (N.P.M.); (S.A.F.)
| | - Jin Han
- Cardiovascular and Metabolic Disease Center, Smart Marine Therapeutic Center, Department of Physiology, College of Medicine, Inje University, Busan 57392, Korea;
- Department of Health Sciences and Technology, Graduate School of Inje University, Busan 57392, Korea
| |
Collapse
|
9
|
Chen C, Zhang M, Zhang Y. Circ_0000079 Decoys the RNA-Binding Protein FXR1 to Interrupt Formation of the FXR1/PRCKI Complex and Decline Their Mediated Cell Invasion and Drug Resistance in NSCLC. Cell Transplant 2021; 29:963689720961070. [PMID: 32951448 PMCID: PMC7784611 DOI: 10.1177/0963689720961070] [Citation(s) in RCA: 20] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Nonsmall cell lung cancer (NSCLC) has gradually become one of the deadliest threats to human health and life worldwide. Although reports have shown that circular RNAs (circRNAs) are associated with progression and metastasis of NSCLC, the biological functions of circRNAs during these processes remain largely unknown. Our study showed that circ_0000079 (CiR79) levels were significantly downregulated in NSCLC patients, especially in cisplatin (DDP)-resistant NSCLC patients, and low circ_0000079 levels were significantly associated with poor overall survival of NSCLC patients. Then, results from Cell Counting Kit-8 (CCK-8) cell viability assay and transwell cell invasion assay in A549/DDP and H460/DDP cells transfected with pCDH-CiR79 expression vector showed that circ_0000079 overexpression significantly inhibited cell proliferation and invasion of these DDP-resistant NSCLC cells. The online bioinformatic program StarBase and RNA-binding protein immunoprecipitation predicted and demonstrated that circ_0000079 could bind with the Fragile X-Related 1 (FXR1) protein rather than with protein kinase C, iota (PRKCI), which was shown to form a complex with FXR1 to promote invasion and growth of NSCLC cells. Co-immunoprecipitation combined with Western blot assays indicated that FXR1 levels were remarkably decreased, but PRKCI levels remained unchanged in pCDH-ciR79 transfected NSCLC cells. Moreover, circ_0000079 negatively regulated FXR1/PRKCI-mediated phosphorylation of glycogen synthesis kinase 3β and activator protein 1, thus suppressing the protein level of the Snail gene, an important promoter gene regulating cancer cell growth and epithelial-mesenchymal transition. Furthermore, DDP resistance of A549/DDP and H460/DDP cells was inhibited by circ_0000079 overexpression but was restored by FXR1. Hence, our findings demonstrated that circ_0000079 might inhibit cell invasion and drug resistance in NSCLC by interrupting the formation of the FXR1/PRCKI complex by interacting with FXR1, and circ_0000079 could act as a potential biomarker and therapeutic target for NSCLC.
Collapse
Affiliation(s)
- Chen Chen
- Department of Pathology, 569063the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Min Zhang
- Department of Pathology, 569063the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| | - Yan Zhang
- Department of Pathology, 569063the Second Affiliated Hospital of Zhengzhou University, Zhengzhou, P. R. China
| |
Collapse
|
10
|
Protein Kinase C as a Therapeutic Target in Non-Small Cell Lung Cancer. Int J Mol Sci 2021; 22:ijms22115527. [PMID: 34073823 PMCID: PMC8197251 DOI: 10.3390/ijms22115527] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2021] [Revised: 05/19/2021] [Accepted: 05/20/2021] [Indexed: 12/30/2022] Open
Abstract
Driver-directed therapeutics have revolutionized cancer treatment, presenting similar or better efficacy compared to traditional chemotherapy and substantially improving quality of life. Despite significant advances, targeted therapy is greatly limited by resistance acquisition, which emerges in nearly all patients receiving treatment. As a result, identifying the molecular modulators of resistance is of great interest. Recent work has implicated protein kinase C (PKC) isozymes as mediators of drug resistance in non-small cell lung cancer (NSCLC). Importantly, previous findings on PKC have implicated this family of enzymes in both tumor-promotive and tumor-suppressive biology in various tissues. Here, we review the biological role of PKC isozymes in NSCLC through extensive analysis of cell-line-based studies to better understand the rationale for PKC inhibition. PKC isoforms α, ε, η, ι, ζ upregulation has been reported in lung cancer, and overexpression correlates with worse prognosis in NSCLC patients. Most importantly, PKC isozymes have been established as mediators of resistance to tyrosine kinase inhibitors in NSCLC. Unfortunately, however, PKC-directed therapeutics have yielded unsatisfactory results, likely due to a lack of specific evaluation for PKC. To achieve satisfactory results in clinical trials, predictive biomarkers of PKC activity must be established and screened for prior to patient enrollment. Furthermore, tandem inhibition of PKC and molecular drivers may be a potential therapeutic strategy to prevent the emergence of resistance in NSCLC.
Collapse
|
11
|
Mitchell SJ, Rosenblatt J. Early mechanical selection of cell extrusion and extrusion signaling in cancer. Curr Opin Cell Biol 2021; 72:36-40. [PMID: 34034216 DOI: 10.1016/j.ceb.2021.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2021] [Revised: 04/19/2021] [Accepted: 04/20/2021] [Indexed: 12/28/2022]
Abstract
Epithelial cells use the process of extrusion to promote cell death while preserving a tight barrier. To extrude, a cell and its neighbors contract actin and myosin circumferentially and basolaterally to seamlessly squeeze it out of the epithelium. Recent research highlights how early apical pulsatile contractions within the extruding cell might orchestrate contraction in three dimensions so that a cell extrudes out apically. Along with apical constrictions, studies of ion channels and mathematical modeling reveal how differential contraction between cells helps select specific cells to extrude. In addition, several studies have offered new insights into pathways that use extrusion to eliminate transformed cells or cause an aberrant form of extrusion that promotes cell invasion.
Collapse
Affiliation(s)
- Saranne J Mitchell
- Biomedical Engineering Department, The University of Utah, Salt Lake City, UT, USA; The Randall Centre for Cell & Molecular Biophysics, Faculty of Life Sciences & Medicine, Schools of Basic & Medical Biosciences and Cancer & Pharmaceutical Sciences, UK
| | - Jody Rosenblatt
- Biomedical Engineering Department, The University of Utah, Salt Lake City, UT, USA; The Randall Centre for Cell & Molecular Biophysics, Faculty of Life Sciences & Medicine, Schools of Basic & Medical Biosciences and Cancer & Pharmaceutical Sciences, UK.
| |
Collapse
|
12
|
Bensen R, Brognard J. New Therapeutic Opportunities for the Treatment of Squamous Cell Carcinomas: A Focus on Novel Driver Kinases. Int J Mol Sci 2021; 22:2831. [PMID: 33799513 PMCID: PMC7999493 DOI: 10.3390/ijms22062831] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/03/2021] [Revised: 03/03/2021] [Accepted: 03/06/2021] [Indexed: 12/12/2022] Open
Abstract
Squamous cell carcinomas of the lung, head and neck, esophagus, and cervix account for more than two million cases of cancer per year worldwide with very few targetable therapies available and minimal clinical improvement in the past three decades. Although these carcinomas are differentiated anatomically, their genetic landscape shares numerous common genetic alterations. Amplification of the third chromosome's distal portion (3q) is a distinguishing genetic alteration in most of these carcinomas and leads to copy-number gain and amplification of numerous oncogenic proteins. This area of the chromosome harbors known oncogenes involved in squamous cell fate decisions and differentiation, including TP63, SOX2, ECT2, and PIK3CA. Furthermore, novel targetable oncogenic kinases within this amplicon include PRKCI, PAK2, MAP3K13, and TNIK. TCGA analysis of these genes identified amplification in more than 20% of clinical squamous cell carcinoma samples, correlating with a significant decrease in overall patient survival. Alteration of these genes frequently co-occurs and is dependent on 3q-chromosome amplification. The dependency of cancer cells on these amplified kinases provides a route toward personalized medicine in squamous cell carcinoma patients through development of small-molecules targeting these kinases.
Collapse
Affiliation(s)
| | - John Brognard
- Laboratory of Cell and Developmental Signaling, Center for Cancer Research, National Cancer Institute, Frederick, MD 21702, USA;
| |
Collapse
|
13
|
Lagoutte E, Villeneuve C, Fraisier V, Krndija D, Deugnier MA, Chavrier P, Rossé C. A new pipeline for pathophysiological analysis of the mammary gland based on organoid transplantation and organ clearing. J Cell Sci 2020; 133:jcs242495. [PMID: 32467329 PMCID: PMC7328142 DOI: 10.1242/jcs.242495] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2019] [Accepted: 05/13/2020] [Indexed: 12/24/2022] Open
Abstract
Recent developments in techniques for tissue clearing and size reduction have enabled optical imaging of whole organs and the study of rare tumorigenic events in vivo The adult mammary gland provides a unique model for investigating physiological or pathological processes such as morphogenesis or epithelial cell dissemination. Here, we establish a new pipeline to study rare cellular events occurring in the mammary gland, by combining orthotopic transplantation of mammary organoids with the uDISCO organ size reduction and clearing method. This strategy allows us to analyze the behavior of individually labeled cells in regenerated mammary gland. As a proof of concept, we analyzed the localization of rare epithelial cells overexpressing atypical protein kinase C iota (also known as PRKCI, referred to here as aPKCι) with an N-terminal eGFP fusion (GFP-aPKCι+) in the normal mammary gland. Using this analytical pipeline, we were able to visualize epithelial aPKCι+ cells escaping from the normal mammary epithelium and disseminating into the surrounding stroma. This technical resource should benefit mammary development and tumor progression studies.
Collapse
Affiliation(s)
- Emilie Lagoutte
- Research center, Institut Curie, Paris Sciences et Lettres Research University, Sorbonne Université, CNRS (Centre National de la Recherche Scientifique), UMR 144, 26 rue d'Ulm, F-75005 Paris, France
| | - Clémentine Villeneuve
- Research center, Institut Curie, Paris Sciences et Lettres Research University, Sorbonne Université, CNRS (Centre National de la Recherche Scientifique), UMR 144, 26 rue d'Ulm, F-75005 Paris, France
| | - Vincent Fraisier
- Research center, Institut Curie, Paris Sciences et Lettres Research University, Sorbonne Université, CNRS (Centre National de la Recherche Scientifique), UMR 144, 26 rue d'Ulm, F-75005 Paris, France
| | - Denis Krndija
- Research center, Institut Curie, Paris Sciences et Lettres Research University, Sorbonne Université, CNRS (Centre National de la Recherche Scientifique), UMR 144, 26 rue d'Ulm, F-75005 Paris, France
| | - Marie-Ange Deugnier
- Research center, Institut Curie, Paris Sciences et Lettres Research University, Sorbonne Université, CNRS (Centre National de la Recherche Scientifique), UMR 144, 26 rue d'Ulm, F-75005 Paris, France
- Institut national de la santé et de la recherche médicale, INSERM, Paris F-75013, France
| | - Philippe Chavrier
- Research center, Institut Curie, Paris Sciences et Lettres Research University, Sorbonne Université, CNRS (Centre National de la Recherche Scientifique), UMR 144, 26 rue d'Ulm, F-75005 Paris, France
| | - Carine Rossé
- Research center, Institut Curie, Paris Sciences et Lettres Research University, Sorbonne Université, CNRS (Centre National de la Recherche Scientifique), UMR 144, 26 rue d'Ulm, F-75005 Paris, France
- Institut national de la santé et de la recherche médicale, INSERM, Paris F-75013, France
| |
Collapse
|
14
|
Transcription co-activator P300 activates Elk1-aPKC-ι signaling mediated epithelial-to-mesenchymal transition and malignancy in hepatocellular carcinoma. Oncogenesis 2020; 9:32. [PMID: 32144235 PMCID: PMC7060348 DOI: 10.1038/s41389-020-0212-5] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/16/2020] [Revised: 01/28/2020] [Accepted: 02/03/2020] [Indexed: 12/20/2022] Open
Abstract
Epithelial-to-mesenchymal transition (EMT) plays an important role in invasion and metastasis of hepatocellular carcinoma (HCC). Our previous study found that atypical protein kinase C-ι (aPKC-ι) promoted the EMT process in HCC. However, how the aPKC-ι signaling pathway is regulated in HCC has not been elucidated. In this study, vector transfection was utilized to study the invasion of HCC cells, and the mechanism between P300 and aPKC-ι signaling pathways in regulating the EMT process of HCC was further elucidated in vitro and in vivo. We found both P300 and aPKC-ι were highly expressed in HCC and they were correlated with tumor progression and poor survival in HCC patients. P300 knockdown inhibited EMT, invasion and other malignant events of HCC cells but promoted cell apoptosis and cycle arrest. However, the effects mediated by P300 knockdown were abolished by aPKC-ι overexpression. Further studies showed that P300 upregulates aPKC-ι expression through increasing the transcription of Elk1, a transcriptional activator of aPKC-ι, and stabilizing Elk1 protein and its phosphorylation. In conclusion, our work uncovered the molecular mechanism by which oncogenic aPKC-ι is upregulated in HCC and suggests that P300, like aPKC-ι, may be used as a prognostic biomarker and therapeutic target in patients with HCC.
Collapse
|
15
|
BommaReddy RR, Patel R, Smalley T, Acevedo-Duncan M. Effects of Atypical Protein Kinase C Inhibitor (DNDA) on Lung Cancer Proliferation and Migration by PKC-ι/FAK Ubiquitination Through the Cbl-b Pathway. Onco Targets Ther 2020; 13:1661-1676. [PMID: 32158232 PMCID: PMC7047975 DOI: 10.2147/ott.s224866] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/29/2019] [Accepted: 02/03/2020] [Indexed: 01/23/2023] Open
Abstract
Purpose The options for treating lung cancers are limited, as diagnosis typically occurs during the late stages of the disease. There is a dire need to develop aPKC (atypical Protein Kinase C) inhibitors due to aPKC overexpression and contributions to lung cancer malignancies. In this study, we investigate the role of atypical PKCs (aPKCs) in cell proliferation and migration in lung cancer cell lines and the effect of the novel aPKC inhibitor DNDA (3,4-amino-2,7 napthalene disulfonic acid). Methods The normal and lung cancer cells were treated with various concentrations of DNDA. We used a WST assay to determine lung cell viability, then analyzed cell apoptosis through Annexin V/PI staining and flow cytometry. Immunoprecipitation determined the proteins' associations, and Western blot allowed testing of the expression of interest proteins. We also employed the UbiTest to identify the ubiquitination of the FAK. The scratch and transwell assays measured cell migration and invasion of lung cancer cells. Results Our data from cell viability and flow cytometry showed a significant reduction in cell proliferation and induction of apoptosis with DNDA treatment in lung cancer cells, as well as no toxic effect on normal BEAS-2B lung cells. Western blot results showed that the phosphorylation of PKC-iota and phosphorylation of FAK decreased in A549 lung cancer cells upon DNDA treatment. Immunoprecipitation (IP) data revealed an association of PKC-ι with FAK and FAK with Casitas B-lineage lymphoma proto-oncogene-b (Cbl-b). UbiTest results suggest that PKC-ι regulates FAK cleavage through its ubiquitination by Cbl-b, thereby inhibiting A549 lung cancer cells' migration. This was evident from scratch, invasion, and migration assays. Conclusion Our study data suggest that DNDA inhibits cell proliferation and induces apoptosis in lung cancer cells. Moreover, DNDA inhibit A549 lung cancer cells' migration by PKC- ι/FAK ubiquitination via Cbl-b.
Collapse
Affiliation(s)
| | - Rekha Patel
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA
| | - Tracess Smalley
- Department of Chemistry, University of South Florida, Tampa, FL 33620, USA
| | | |
Collapse
|
16
|
aPKCi triggers basal extrusion of luminal mammary epithelial cells by tuning contractility and vinculin localization at cell junctions. Proc Natl Acad Sci U S A 2019; 116:24108-24114. [PMID: 31699818 PMCID: PMC6883778 DOI: 10.1073/pnas.1906779116] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
This study shows that an oncogenic mammary epithelial cell surrounded by normal cells can extrude basally in vivo and invade surrounding tissues without formation of a primary tumor. Here, we show that overexpression of the key polarity protein atypical protein kinase C ι (aPKCi) is sufficient for triggering basally oriented epithelial cell extrusion and early cell invasion into the mammary gland stroma. Moreover, we highlight the importance of the difference between the mechanical properties of aPKCi-overexpressing cells and those of the normal surrounding cells associated with the decrease of vinculin at the cell junction, which triggers cell segregation, the first step toward promoting and controlling the direction of cell extrusion. Metastasis is the main cause of cancer-related deaths. How a single oncogenic cell evolves within highly organized epithelium is still unknown. Here, we found that the overexpression of the protein kinase atypical protein kinase C ι (aPKCi), an oncogene, triggers basally oriented epithelial cell extrusion in vivo as a potential mechanism for early breast tumor cell invasion. We found that cell segregation is the first step required for basal extrusion of luminal cells and identify aPKCi and vinculin as regulators of cell segregation. We propose that asymmetric vinculin levels at the junction between normal and aPKCi+ cells trigger an increase in tension at these cell junctions. Moreover, we show that aPKCi+ cells acquire promigratory features, including increased vinculin levels and vinculin dynamics at the cell–substratum contacts. Overall, this study shows that a balance between cell contractility and cell–cell adhesion is crucial for promoting basally oriented cell extrusion, a mechanism for early breast cancer cell invasion.
Collapse
|
17
|
Yin N, Liu Y, Murray NR, Fields AP. Oncogenic protein kinase Cι signaling mechanisms in lung cancer: Implications for improved therapeutic strategies. Adv Biol Regul 2019; 75:100656. [PMID: 31623973 DOI: 10.1016/j.jbior.2019.100656] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/03/2019] [Revised: 09/19/2019] [Accepted: 09/21/2019] [Indexed: 11/28/2022]
Abstract
Protein Kinase Cι (PKCι) is a major oncogene involved in the initiation, maintenance and progression of numerous forms of human cancer. In the lung, PKCι is necessary for the maintenance of the transformed phenotype of the two major forms of non-small cell lung cancer (NSCLC), lung adenocarcinoma (LADC) and lung squamous cell carcinoma (LSCC). In addition, PKCι is necessary for both LADC and LSCC tumorigenesis by establishing and maintaining a highly aggressive stem-like, tumor-initiating cell phenotype. Interestingly however, while PKCι signaling in these two major lung cancer subtypes shares some common elements, it also drives distinct, sub-type specific pathways. Furthermore, recent analysis has revealed both PKCι-dependent and PKCι-independent pathways to LADC development. Herein, we discussion our current knowledge of oncogenic PKCι signaling in LADC and LSCC, and discuss these findings in the context of how they may inform strategies for improved therapeutic intervention in these deadly diseases.
Collapse
Affiliation(s)
- Ning Yin
- From the Department of Cancer Cell Biology, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | - Yi Liu
- From the Department of Cancer Cell Biology, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | - Nicole R Murray
- From the Department of Cancer Cell Biology, Mayo Clinic Florida, Jacksonville, FL, 32224, USA
| | - Alan P Fields
- From the Department of Cancer Cell Biology, Mayo Clinic Florida, Jacksonville, FL, 32224, USA.
| |
Collapse
|
18
|
Liu L, Lei B, Wang L, Chang C, Yang H, Liu J, Huang G, Xie W. Protein kinase C-iota-mediated glycolysis promotes non-small-cell lung cancer progression. Onco Targets Ther 2019; 12:5835-5848. [PMID: 31410027 PMCID: PMC6646854 DOI: 10.2147/ott.s207211] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2019] [Accepted: 06/18/2019] [Indexed: 12/02/2022] Open
Abstract
Purpose To determine whether protein kinase C-iota (PKC-iota) is associated with glucose metabolism in non-small-cell lung cancer (NSCLC) and whether its regulatory effect on metabolic and biological changes observed in NSCLC can be mediated by glucose transporter 1 (GLUT1). Patients and methods Forty-five NSCLC patients underwent combined 18F-fludeoxyglucose (18F-FDG) positron emission tomography and computed tomography (PET/CT) before surgery, and another eighty-one NSCLC patients were followed-up for 1–91 months after tumor resection. The rate of glucose metabolism in NSCLC was quantified by measuring the maximum standardized uptake value (SUVmax) by 18F-FDG PET/CT. PKC-iota and GLUT1 in NSCLC were detected by immunostaining. In vitro, PKC-iota was knocked down, whereas GLUT1 was silenced with or without PKC-iota overexpression to identify the role of PKC-iota in glycolysis. Spearman’s rank correlation coefficient was used in the correlation analysis. Kaplan-Meier analysis was used to assess survival duration. Results There was a positive relationship between PKC-iota expression and SUVmax in NSCLC (r=0.649, P<0.001). PKC-iota expression also showed a positive relationship with GLUT1 in NSCLC tissues (r=0.686, P<0.001). Patients whose NSCLC tissues highly co-expressed PKC-iota and GLUT1 had worse prognosis compared with patients without high co-expression of PKC-iota and GLUT1. In vitro, PKC-iota silencing significantly decreased the expression of GLUT1 and inhibited glucose uptake and glycolysis; c-Myc silencing restrained PKC-iota-mediated GLUT1 elevation; GLUT1 knockdown remarkably suppressed PKC-iota-mediated glycolysis and cell growth. Conclusion In NSCLC, the rate of glucose metabolism was positively correlated with PKC-iota expression. PKC-iota increased glucose accumulation and glycolysis by upregulating c-Myc/GLUT1 signaling and is thus involved in tumor progression.
Collapse
Affiliation(s)
- Liu Liu
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Bei Lei
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Lihua Wang
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Cheng Chang
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Hao Yang
- Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, People's Republic of China
| | - Jianjun Liu
- Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Gang Huang
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China.,Shanghai Key Laboratory for Molecular Imaging, Shanghai University of Medicine and Health Sciences, Shanghai, People's Republic of China.,Department of Nuclear Medicine, Renji Hospital, School of Medicine, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| | - Wenhui Xie
- Department of Nuclear Medicine, Shanghai Chest Hospital, Shanghai Jiao Tong University, Shanghai, People's Republic of China
| |
Collapse
|
19
|
Kwiatkowski J, Baburajendran N, Poulsen A, Liu B, Tee DHY, Wong YX, Poh ZY, Ong EHQ, Dinie N, Cherian J, Jansson AE, Hill J, Keller TH, Hung AW. Fragment-based Discovery of a Small-Molecule Protein Kinase C-iota Inhibitor Binding Post-kinase Domain Residues. ACS Med Chem Lett 2019; 10:318-323. [PMID: 30891133 DOI: 10.1021/acsmedchemlett.8b00546] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2018] [Accepted: 02/15/2019] [Indexed: 02/02/2023] Open
Abstract
The atypical protein kinase C-iota (PKC-ι) enzyme is implicated in various cancers and has been put forward as an attractive target for developing anticancer therapy. A high concentration biochemical screen identified pyridine fragment weakly inhibiting PKC-ι with IC50 = 424 μM. Driven by structure-activity relationships and guided by docking hypothesis, the weakly bound fragment was eventually optimized into a potent inhibitor of PKC-ι (IC50= 270 nM). Through the course of the optimization, an intermediate compound was crystallized with the protein, and careful analysis of the X-ray crystal structure revealed a unique binding mode involving the post-kinase domain (C-terminal tail) of PKC-ι.
Collapse
Affiliation(s)
- Jacek Kwiatkowski
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios #03-10/11, Singapore 138667, Singapore
| | - Nithya Baburajendran
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios #03-10/11, Singapore 138667, Singapore
| | - Anders Poulsen
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios #03-10/11, Singapore 138667, Singapore
| | - Boping Liu
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios #03-10/11, Singapore 138667, Singapore
| | - Doris Hui Ying Tee
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios #03-10/11, Singapore 138667, Singapore
| | - Yun Xuan Wong
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios #03-10/11, Singapore 138667, Singapore
| | - Zhi Ying Poh
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios #03-10/11, Singapore 138667, Singapore
| | - Esther HQ Ong
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios #03-10/11, Singapore 138667, Singapore
| | - Nurul Dinie
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios #03-10/11, Singapore 138667, Singapore
| | - Joseph Cherian
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios #03-10/11, Singapore 138667, Singapore
| | - Anna Elisabet Jansson
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios #03-10/11, Singapore 138667, Singapore
| | - Jeffrey Hill
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios #03-10/11, Singapore 138667, Singapore
| | - Thomas H. Keller
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios #03-10/11, Singapore 138667, Singapore
| | - Alvin W. Hung
- Experimental Therapeutics Centre, Agency for Science, Technology and Research (A*STAR), 11 Biopolis Way, Helios #03-10/11, Singapore 138667, Singapore
| |
Collapse
|
20
|
Cai X, Zhu H, Li Y. PKCζ, MMP‑2 and MMP‑9 expression in lung adenocarcinoma and association with a metastatic phenotype. Mol Med Rep 2017; 16:8301-8306. [PMID: 28983601 DOI: 10.3892/mmr.2017.7634] [Citation(s) in RCA: 25] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2017] [Accepted: 08/01/2017] [Indexed: 11/06/2022] Open
Abstract
The aim of the present study was to investigate protein kinase C ζ type (PKCζ), matrix metalloproteinase (MMP)‑2 and MMP‑9 expression in lung adenocarcinoma and to define their association with in vitro invasion and metastatic capacity. PKCζ, MMP‑2 and MMP‑9 expression was assessed by immunohistochemistry in 110 cases of lung adenocarcinoma. PKCζ small interfering (si)RNA was transfected into A549 cells, and western blotting was used to confirm PKCζ‑knockdown in transfected cells and to measure MMP‑2 and MMP‑9 levels. A Transwell invasion assay was used to detect in vitro invasive capacity. The rates of positive PKCζ, MMP‑2 and MMP‑9 staining in lung adenocarcinoma tissues were 52.73, 55.45 and 61.82%, respectively. PKCζ expression was increased in malignant tissues compared with adjacent normal lung tissues and was associated with lymph node metastasis (P<0.05), although it was not associated with any other clinicopathological parameters, including sex, age, tumor size, smoking status or distant metastases (all P>0.05). PKCζ, MMP‑2 and MMP‑9 expression was markedly decreased in siPKCζ‑treated A549 cells, which exhibited a significantly decreased invasive capacity in the Transwell invasion assay (P<0.05). In conclusion, PKCζ promoted lung adenocarcinoma invasion and metastasis, and its expression was associated with MMP‑2 and MMP‑9 expression. PKCζ may be a potential target for gene therapy in lung adenocarcinoma.
Collapse
Affiliation(s)
- Xiaoshan Cai
- Department of Pathology, Second People's Hospital of Weifang, Weifang, Shandong 261041, P.R. China
| | - Hongguang Zhu
- Department of Dentistry, Weifang People's Hospital, Weifang, Shandong 261000, P.R. China
| | - Ying Li
- Department of Pathology, Second People's Hospital of Weifang, Weifang, Shandong 261041, P.R. China
| |
Collapse
|
21
|
Yeo MK, Kim JY, Seong IO, Kim JM, Kim KH. Phosphorylated Protein Kinase C (Zeta/Lambda) Expression in Colorectal Adenocarcinoma and Its Correlation with Clinicopathologic Characteristics and Prognosis. J Cancer 2017; 8:3371-3377. [PMID: 29158810 PMCID: PMC5665054 DOI: 10.7150/jca.20983] [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: 05/11/2017] [Accepted: 08/29/2017] [Indexed: 12/29/2022] Open
Abstract
Background: Protein kinase C zeta/lambda (PKCζ/λ) is a family of protein kinase enzymes that contributes to cell proliferation and regulation, which are important for cancer development. PKCζ/λ has been shown to be an important regulator of tumorigenesis in intestinal cancer. The phosphorylated form of PKCζ/λ, p-PKCζ/λ, is suggested as an active form of PKCζ/λ. However, p-PKCζ/λ expression and its clinicopathologic implication in colorectal adenocarcinoma (CRAC) are unclear. Methods: Seven whole-tissue sections of malignant polyps containing both non-neoplastic and neoplastic mucosa, 11 adenomas with low-grade dysplasia, and 173 CRACs were examined by immunohistochemistry and western blot assay for p-PKCζ/λ protein expression. The association of p-PKCζ/λ expression with clinicopathologic factors including patient survival was studied. Results: In non-neoplastic epithelia, p-PKCζ/λ showed a weak cytoplasmic immunostaining. Adenomas and CRACs demonstrated up-regulated p-PKCζ/λ detection. Cytoplasmic p-PKCζ/λ expression was higher in CRAC than in adenoma. In CRACs, p-PKCζ/λ expression was inversely correlated with pathologic TNM stage (I-II versus III-IV) and poor differentiation. Statistical correlations between low expression of p-PKCζ/λ with shortened overall survival and disease-free survival were seen (p=0.004 and p=0.034, respectively). Conclusions: P-PKCζ/λ overexpression is implicated in tumorigenesis but down-regulation was a poor prognostic factor in CRAC.
Collapse
Affiliation(s)
- Min-Kyung Yeo
- Department of Pathology, Cancer Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Ji Yeon Kim
- Department of Surgery, Chungnam National University Hospital, Daejeon, Republic of Korea
| | - In-Ock Seong
- Department of Pathology, Cancer Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Jin-Man Kim
- Department of Pathology and Medical Science, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| | - Kyung-Hee Kim
- Department of Pathology, Cancer Research Institute, Chungnam National University School of Medicine, Daejeon, Republic of Korea
| |
Collapse
|
22
|
Davidson MA, Shanks EJ. 3q26-29 Amplification in head and neck squamous cell carcinoma: a review of established and prospective oncogenes. FEBS J 2017; 284:2705-2731. [PMID: 28317270 DOI: 10.1111/febs.14061] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2016] [Revised: 02/23/2017] [Accepted: 03/15/2017] [Indexed: 12/22/2022]
Abstract
Head and neck squamous cell carcinoma (HNSCC) is significantly underrepresented in worldwide cancer research, yet survival rates for the disease have remained static for over 50 years. Distant metastasis is often present at the time of diagnosis, and is the primary cause of death in cancer patients. In the absence of routine effective targeted therapies, the standard of care treatment remains chemoradiation in combination with (often disfiguring) surgery. A defining characteristic of HNSCC is the amplification of a region of chromosome 3 (3q26-29), which is consistently associated with poorer patient outcome. This review provides an overview of the role the 3q26-29 region plays in HNSCC, in terms of both known and as yet undiscovered processes, which may have potential clinical relevance.
Collapse
|
23
|
Hua K, Lin CH, Chen YL, Lin CH, Ping YH, Jou YS, Chen CF. Identification of novel cancer fusion genes using chromosome breakpoint screening. Oncol Rep 2017; 37:2101-2108. [DOI: 10.3892/or.2017.5492] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2016] [Accepted: 09/22/2016] [Indexed: 11/06/2022] Open
|
24
|
Abstract
Atypical protein kinase Cι (PKCι) is an oncogene in lung and ovarian cancer. The PKCι gene PRKCI is targeted for frequent tumor-specific copy number gain (CNG) in both lung squamous cell carcinoma (LSCC) and ovarian serous carcinoma (OSC). We recently demonstrated that in LSCC cells PRKCI CNG functions to drive transformed growth and tumorigenicity by activating PKCι-dependent cell autonomous Hedgehog (Hh) signaling. Here, we assessed whether OSC cells harboring PRKCI CNG exhibit similar PKCι-dependent Hh signaling. Surprisingly, we find that whereas PKCι is required for the transformed growth of OSC cells harboring PRKCI CNG, these cells do not exhibit PKCι-dependent Hh signaling or Hh-dependent proliferation. Rather, transformed growth of OSC cells is regulated by PKCι-dependent nuclear localization of the oncogenic transcription factor, YAP1. Lentiviral shRNA-mediated knockdown (KD) of PKCι leads to decreased nuclear YAP1 and increased YAP1 binding to angiomotin (AMOT), which sequesters YAP1 in the cytoplasm. Biochemical analysis reveals that PKCι directly phosphorylates AMOT at a unique site, Thr750, whose phosphorylation inhibits YAP1 binding. Pharmacologic inhibition of PKCι decreases YAP1 nuclear localization and blocks OSC tumor growth in vitro and in vivo. Immunohistochemical analysis reveals a strong positive correlation between tumor PKCι expression and nuclear YAP1 in primary OSC tumor samples, indicating the clinical relevance of PKCι-YAP1 signaling. Our results uncover a novel PKCι-AMOT-YAP1 signaling axis that promotes OSC tumor growth, and provide a rationale for therapeutic targeting of this pathway for treatment of OSC.
Collapse
|
25
|
Prkci is required for a non-autonomous signal that coordinates cell polarity during cavitation. Dev Biol 2016; 416:82-97. [PMID: 27312576 DOI: 10.1016/j.ydbio.2016.06.002] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2015] [Revised: 05/26/2016] [Accepted: 06/01/2016] [Indexed: 11/23/2022]
Abstract
Polarized epithelia define boundaries, spaces, and cavities within organisms. Cavitation, a process by which multicellular hollow balls or tubes are produced, is typically associated with the formation of organized epithelia. In order for these epithelial layers to form, cells must ultimately establish a distinct apical-basal polarity. Atypical PKCs have been proposed to be required for apical-basal polarity in diverse species. Here we show that while cells null for the Prkci isozyme exhibit some polarity characteristics, they fail to properly segregate apical-basal proteins, form a coordinated ectodermal epithelium, or participate in normal cavitation. A failure to cavitate could be due to an overgrowth of interior cells or to an inability of interior cells to die. Null cells however, do not have a marked change in proliferation rate and are still capable of undergoing cell death, suggesting that alterations in these processes are not the predominant cause of the failed cavitation. Overexpression of BMP4 or EZRIN can partially rescue the phenotype possibly by promoting cell death, polarity, and differentiation. However, neither is sufficient to provide the required cues to generate a polarized epithelium and fully rescue cavitation. Interestingly, when wildtype and Prkci(-/-) ES cells are mixed together, a polarized ectodermal epithelium forms and cavitation is rescued, likely due to the ability of wildtype cells to produce non-autonomous polarity cues. We conclude that Prkci is not required for cells to respond to these cues, though it is required to produce them. Together these findings indicate that environmental cues can facilitate the formation of polarized epithelia and that cavitation requires the proper coordination of multiple basic cellular processes including proliferation, differentiation, cell death, and apical-basal polarization.
Collapse
|
26
|
Wang Y, Justilien V, Brennan KI, Jamieson L, Murray NR, Fields AP. PKCι regulates nuclear YAP1 localization and ovarian cancer tumorigenesis. Oncogene 2016; 36:534-545. [PMID: 27321186 PMCID: PMC5173453 DOI: 10.1038/onc.2016.224] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/12/2016] [Revised: 05/06/2016] [Accepted: 05/15/2016] [Indexed: 01/08/2023]
Abstract
Atypical protein kinase Cι (PKCι) is an oncogene in lung and ovarian cancer. The PKCι gene PRKCI is targeted for frequent tumor-specific copy number gain (CNG) in both lung squamous cell carcinoma (LSCC) and ovarian serous carcinoma (OSC). We recently demonstrated that in LSCC cells PRKCI CNG functions to drive transformed growth and tumorigenicity by activating PKCι-dependent cell autonomous Hedgehog (Hh) signaling. Here, we assessed whether OSC cells harboring PRKCI CNG exhibit similar PKCι-dependent Hh signaling. Surprisingly, we find that whereas PKCι is required for the transformed growth for OSC cells harboring PRKCI CNG, these cells do not exhibit PKCι-dependent Hh signaling or Hh-dependent proliferation. Rather, transformed growth of OSC cells is regulated by PKCι-dependent nuclear localization of the oncogenic transcription factor, YAP1. Lentiviral shRNA-mediated knock down (KD) of PKCι leads to decreased nuclear YAP1 and increased YAP1 binding to angiomotin (AMOT), which sequesters YAP1 in the cytoplasm. Biochemical analysis reveals that PKCι directly phosphorylates AMOT at a unique site, Thr750, whose phosphorylation inhibits YAP1 binding. Pharmacologic inhibition of PKCι decreases YAP1 nuclear localization and blocks OSC tumor growth in vitro and in vivo. Immunohistochemical analysis reveals a strong positive correlation between tumor PKCι expression and nuclear YAP1 in primary OSC tumor samples, indicating the clinical relevance of PKCι-YAP1 signaling. Our results uncover a novel PKCι-AMOT-YAP1 signaling axis that promotes OSC tumor growth, and provide a rationale for therapeutic targeting of this pathway for treatment of OSC.
Collapse
Affiliation(s)
- Y Wang
- Department of Cancer Biology, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - V Justilien
- Department of Cancer Biology, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - K I Brennan
- Department of Cancer Biology, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - L Jamieson
- Department of Cancer Biology, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - N R Murray
- Department of Cancer Biology, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| | - A P Fields
- Department of Cancer Biology, Mayo Clinic College of Medicine, Jacksonville, FL, USA
| |
Collapse
|
27
|
Fields AP, Ali SA, Justilien V, Murray NR. Targeting oncogenic protein kinase Cι for treatment of mutant KRAS LADC. Small GTPases 2016; 8:58-64. [PMID: 27245608 DOI: 10.1080/21541248.2016.1194953] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Abstract
Lung cancer is the leading cause of cancer death in the US with ∼124,000 new cases annually, and a 5 y survival rate of ∼16%. Mutant KRAS-driven lung adenocarcinoma (KRAS LADC) is a particularly prevalent and deadly form of lung cancer. Protein kinase Cι (PKCι) is an oncogenic effector of KRAS that activates multiple signaling pathways that stimulate transformed growth and invasion, and maintain a KRAS LADC tumor-initiating cell (TIC) phenotype. PKCι inhibitors used alone and in strategic combination show promise as new therapeutic approaches to treatment of KRAS LADC. These novel drug combinations may improve clinical management of KRAS LADC.
Collapse
Affiliation(s)
- Alan P Fields
- a Department of Cancer Biology , Mayo Clinic , Jacksonville , FL , USA
| | - Syed A Ali
- a Department of Cancer Biology , Mayo Clinic , Jacksonville , FL , USA
| | - Verline Justilien
- a Department of Cancer Biology , Mayo Clinic , Jacksonville , FL , USA
| | - Nicole R Murray
- a Department of Cancer Biology , Mayo Clinic , Jacksonville , FL , USA
| |
Collapse
|
28
|
Gunaratne A, Chan E, El-Chabib TH, Carter D, Di Guglielmo GM. aPKC alters the TGFβ response in NSCLC cells through both Smad-dependent and Smad-independent pathways. J Cell Sci 2016; 128:487–98. [PMID: 25501807 DOI: 10.1242/jcs.155440] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/29/2023] Open
Abstract
Transforming growth factor b (TGFb) signaling controls many cellular responses including proliferation, epithelial to mesenchymal transition and apoptosis, through the activation of canonical (Smad) as well as non-canonical (e.g., Par6) pathways. Previous studies from our lab have demonstrated that aPKC inhibition regulates TGFb receptor trafficking and signaling. Here, we report that downstream TGFb-dependent transcriptional responses in aPKC-silenced NSCLC cells were reduced compared with those of control cells, despite a temporal extension of Smad2 phosphorylation. We assessed SARA–Smad2–Smad4 association and observed that knockdown of aPKC increased SARA (also known as ZFYVE9) levels and SARA–Smad2 complex formation, increased cytoplasmic retention of Smad2 and reduced Smad2–Smad4 complex formation, which correlated with reduced Smad2 nuclear translocation. Interestingly, we also detected an increase in p38 MAPK phosphorylation and apoptosis in aPKC-silenced cells, which were found to be TRAF6-dependent. Taken together, our results suggest that aPKC isoforms regulate Smad and non-Smad TGFb pathways and that aPKC inhibition sensitizes NSCLC cells to undergo TGFb dependent apoptosis.
Collapse
|
29
|
Glycogen Synthase Kinase 3β Is Positively Regulated by Protein Kinase Cζ-Mediated Phosphorylation Induced by Wnt Agonists. Mol Cell Biol 2015; 36:731-41. [PMID: 26711256 DOI: 10.1128/mcb.00828-15] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/27/2015] [Accepted: 12/04/2015] [Indexed: 02/01/2023] Open
Abstract
The molecular events that drive Wnt-induced regulation of glycogen synthase kinase 3β (GSK-3β) activity are poorly defined. In this study, we found that protein kinase Cζ (PKCζ) and GSK-3β interact mainly in colon cancer cells. Wnt stimulation induced a rapid GSK-3β redistribution from the cytoplasm to the nuclei in malignant cells and a transient PKC-mediated phosphorylation of GSK-3β at a different site from serine 9. In addition, while Wnt treatment induced a decrease in PKC-mediated phosphorylation of GSK-3β in nonmalignant cells, in malignant cells, this phosphorylation was increased. Pharmacological inhibition and small interfering RNA (siRNA)-mediated silencing of PKCζ abolished all of these effects, but unexpectedly, it also abolished the constitutive basal activity of GSK-3β. In vitro activity assays demonstrated that GSK-3β phosphorylation mediated by PKCζ enhanced GSK-3β activity. We mapped Ser147 of GSK-3β as the site phosphorylated by PKCζ, i.e., its mutation into alanine abolished GSK-3β activity, resulting in β-catenin stabilization and increased transcriptional activity, whereas phosphomimetic replacement of Ser147 by glutamic acid maintained GSK-3β basal activity. Thus, we found that PKCζ phosphorylates GSK-3β at Ser147 to maintain its constitutive activity in resting cells and that Wnt stimulation modifies the phosphorylation of Ser147 to regulate GSK-3β activity in opposite manners in normal and malignant colon cells.
Collapse
|
30
|
Fields AP, Justilien V, Murray NR. The chromosome 3q26 OncCassette: A multigenic driver of human cancer. Adv Biol Regul 2015; 60:47-63. [PMID: 26754874 DOI: 10.1016/j.jbior.2015.10.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2015] [Revised: 10/28/2015] [Accepted: 10/29/2015] [Indexed: 02/06/2023]
Abstract
Recurrent copy number variations (CNVs) are genetic alterations commonly observed in human tumors. One of the most frequent CNVs in human tumors involves copy number gains (CNGs) at chromosome 3q26, which is estimated to occur in >20% of human tumors. The high prevalence and frequent occurrence of 3q26 CNG suggest that it drives the biology of tumors harboring this genetic alteration. The chromosomal region subject to CNG (the 3q26 amplicon) spans from chromosome 3q26 to q29, a region containing ∼200 protein-encoding genes. The large number of genes within the amplicon makes it difficult to identify relevant oncogenic target(s). Whereas a number of genes in this region have been linked to the transformed phenotype, recent studies indicate a high level of cooperativity among a subset of frequently amplified 3q26 genes. Here we use a novel bioinformatics approach to identify potential driver genes within the recurrent 3q26 amplicon in lung squamous cell carcinoma (LSCC). Our analysis reveals a set of 35 3q26 amplicon genes that are coordinately amplified and overexpressed in human LSCC tumors, and that also map to a major LSCC susceptibility locus identified on mouse chromosome 3 that is syntenic with human chromosome 3q26. Pathway analysis reveals that 21 of these genes exist within a single predicted network module. Four 3q26 genes, SOX2, ECT2, PRKCI and PI3KCA occupy the hub of this network module and serve as nodal genes around which the network is organized. Integration of available genetic, genomic, biochemical and functional data demonstrates that SOX2, ECT2, PRKCI and PIK3CA are cooperating oncogenes that function within an integrated cell signaling network that drives a highly aggressive, stem-like phenotype in LSCC tumors harboring 3q26 amplification. Based on the high level of genomic, genetic, biochemical and functional integration amongst these 4 3q26 nodal genes, we propose that they are the key oncogenic targets of the 3q26 amplicon and together define a "3q26 OncCassette" that mediates 3q26 CNG-driven tumorigenesis. Genomic analysis indicates that the 3q26 OncCassette also operates in other major tumor types that exhibit frequent 3q26 CNGs, including head and neck squamous cell carcinoma (HNSCC), ovarian serous cancer and cervical cancer. Finally, we discuss how the 3q26 OncCassette represents a tractable target for development of novel therapeutic intervention strategies that hold promise for improving treatment of 3q26-driven cancers.
Collapse
Affiliation(s)
- Alan P Fields
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, United States.
| | - Verline Justilien
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, United States
| | - Nicole R Murray
- Department of Cancer Biology, Mayo Clinic Comprehensive Cancer Center, Jacksonville, FL, United States
| |
Collapse
|
31
|
Mah IK, Soloff R, Hedrick SM, Mariani FV. Atypical PKC-iota Controls Stem Cell Expansion via Regulation of the Notch Pathway. Stem Cell Reports 2015; 5:866-880. [PMID: 26527382 PMCID: PMC4649379 DOI: 10.1016/j.stemcr.2015.09.021] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2015] [Revised: 09/25/2015] [Accepted: 09/28/2015] [Indexed: 12/16/2022] Open
Abstract
The number of stem/progenitor cells available can profoundly impact tissue homeostasis and the response to injury or disease. Here, we propose that an atypical PKC, Prkci, is a key player in regulating the switch from an expansion to a differentiation/maintenance phase via regulation of Notch, thus linking the polarity pathway with the control of stem cell self-renewal. Prkci is known to influence symmetric cell division in invertebrates; however a definitive role in mammals has not yet emerged. Using a genetic approach, we find that loss of Prkci results in a marked increase in the number of various stem/progenitor cells. The mechanism used likely involves inactivation and symmetric localization of NUMB, leading to the activation of NOTCH1 and its downstream effectors. Inhibition of atypical PKCs may be useful for boosting the production of pluripotent stem cells, multipotent stem cells, or possibly even primordial germ cells by promoting the stem cell/progenitor fate. PRKCi, a polarity protein, regulates expansion of various stem/progenitor cells PRKCi acts in this capacity via a Notch-dependent pathway Thus, PRKCi acts as a link between polarity and stem cell self-renewal Inhibition of aPKCs may be generally useful for expanding progenitor populations
Collapse
Affiliation(s)
- In Kyoung Mah
- Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, 1425 San Pablo St., Los Angeles, CA 90033, USA
| | - Rachel Soloff
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Stephen M Hedrick
- Department of Biological Sciences, University of California, San Diego, La Jolla, CA 92093, USA
| | - Francesca V Mariani
- Broad CIRM Center for Regenerative Medicine and Stem Cell Research, Keck School of Medicine, University of Southern California, 1425 San Pablo St., Los Angeles, CA 90033, USA.
| |
Collapse
|
32
|
Jin X, Zhai B, Fang T, Guo X, Xu L. FXR1 is elevated in colorectal cancer and acts as an oncogene. Tumour Biol 2015; 37:2683-90. [DOI: 10.1007/s13277-015-4068-9] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2015] [Accepted: 09/13/2015] [Indexed: 01/28/2023] Open
|
33
|
miRNA-mRNA Interaction Network in Non-small Cell Lung Cancer. Interdiscip Sci 2015; 8:209-19. [PMID: 26338522 DOI: 10.1007/s12539-015-0117-8] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/03/2015] [Accepted: 02/09/2015] [Indexed: 12/31/2022]
Abstract
MicroRNAs (miRNAs) are small RNA molecules, about 20-25 nucleotides in length. They repress or degrade messenger RNA (mRNA) translation, which are involved in human cancer. In this study based on paired miRNA and mRNA expression profiles of non-small cell lung cancer samples, we constructed and analyzed miRNA-mRNA interaction network via several bioinformatics softwares and platforms. This integrative network is comprised of 249 nodes for mRNA, 90 nodes for miRNA and 290 edges that show regulations between target genes and miRNAs. The three miR-1207-5p, miR-1228* and miR-939 are the most connected miRNA that regulated a large number of genes. ST8SIA2, MED1 and HDAC4, SPN, which are targeted by multiple miRNAs and located in the center of the network, are involved in both lung cancer and nervous system via functional annotation analysis. Such a global interaction network of miRNA-mRNA in lung cancer will contribute to refining miRNA target predictions and developing novel therapeutic candidates.
Collapse
|
34
|
PKCiota promotes ovarian tumor progression through deregulation of cyclin E. Oncogene 2015; 35:2428-40. [PMID: 26279297 PMCID: PMC4856585 DOI: 10.1038/onc.2015.301] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2015] [Revised: 06/18/2015] [Accepted: 06/29/2015] [Indexed: 01/29/2023]
Abstract
The high frequency of relapse of epithelial ovarian tumors treated with standard chemotherapy has highlighted the necessity to identify targeted therapies that can improve patient outcomes. The dynamic relationship between Cyclin E and PKCiota frequent overexpression in high-grade ovarian tumors poses a novel pathway for therapeutic investigation. We hypothesized that a PI3K dependent signaling pathway activating PKCiota perpetuates cyclin E deregulation during ovarian tumorigenesis. We observed a positive correlation between PKCiota and cyclin E in a panel of 19 ovarian cancer cell lines. Modulation of cyclin E had no effect on PKCiota knockdown/overexpression however PKCiota differentially regulated cyclin E expression. In the serous ovarian cancer cells (IGROV, OVCAR-3), shPKCiota decreased proliferation, caused a G1 arrest, and significantly prolonged overall survival in xenograft mouse models. In vitro shPKCiota decreased the ability of IGROV cells to grow under anchorage independent conditions and form aberrant acini, which was dependent upon Ad-cyclin E or Ad-LMW-E expression. RPPA analysis of PKCiota wild-type, catalytic active, dominant negative protein isoforms strengthened the association between phospho-PKCiota levels and PI3K pathway activation. Inhibitors of PI3K coordinately decreased phospho-PKCiota and Cyclin E protein levels. In conclusion, we have identified a PI3K/PKCiota/Cyclin E signaling pathway as a therapeutic target during ovarian tumorigenesis.
Collapse
|
35
|
Li Q, Gu C, Zhu Y, Wang M, Yang Y, Wang J, Jin L, Zhu ML, Shi TY, He J, Ye D, Wei Q. Two novel PRKCI polymorphisms and prostate cancer risk in an Eastern Chinese Han population. Mol Carcinog 2015; 54:632-41. [PMID: 24510606 DOI: 10.1002/mc.22130] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2013] [Revised: 12/05/2013] [Accepted: 12/19/2013] [Indexed: 02/05/2023]
Abstract
The atypical protein kinase C (aPKCι), encoded by the PRKCI gene, has been recently found to be a unique human oncoprotein, compared with some other diverse PKC isozymes. Genetic variations in PRKCI have also been reported to be associated with prostate cancer (PCa) risk in Caucasian populations, but no similar studies have been reported for Chinese populations. We genotyped two well-described PRKCI single nucleotide polymorphisms (SNPs) rs546950 and rs4955720 in 1015 PCa patients and 1044 cancer-free controls of Eastern Chinese men. SNPs in the vicinity of those two variants of PRKCI were evaluated using the in silico analysis. Logistic regression was then used to estimate their associations with and interactions in PCa risk. Although no significant main effects were found for the two tested SNPs in the single locus analysis, individuals carrying homozygote wide-type form of these two SNPs had slightly reduced PCa risk (adjusted OR = 0.63, 95% CI = 0.40-0.99, P = 0.045), compared with those carrying any of heterozygous or homozygous variant genotypes. Our results indicated that the two PRKCI SNPs were jointly associated with PCa risk in an Eastern Chinese population. Larger studies with multiethnic groups are warranted to confirm these findings and to explore the role of PRKCI SNPs in the etiology of PCa.
Collapse
Affiliation(s)
- Qiaoxin Li
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Chengyuan Gu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Yao Zhu
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Mengyun Wang
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Yajun Yang
- Ministry of Education Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Jiucun Wang
- Ministry of Education Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Li Jin
- Ministry of Education Key Laboratory of Contemporary Anthropology and State Key Laboratory of Genetic Engineering, School of Life Sciences, Fudan University, Shanghai, China
- Fudan-Taizhou Institute of Health Sciences, Taizhou, Jiangsu, China
| | - Mei-Ling Zhu
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Ting-Yan Shi
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Jing He
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
| | - Dingwei Ye
- Department of Oncology, Shanghai Medical College, Fudan University, Shanghai, China
- Department of Urology, Fudan University Shanghai Cancer Center, Shanghai, China
| | - Qingyi Wei
- Cancer Institute, Fudan University Shanghai Cancer Center, Shanghai, China
- Duke Cancer Institute, Duke University Medical Center, Durham, North Carolina
| |
Collapse
|
36
|
PKCζ Promotes Breast Cancer Invasion by Regulating Expression of E-cadherin and Zonula Occludens-1 (ZO-1) via NFκB-p65. Sci Rep 2015. [PMID: 26218882 PMCID: PMC4648478 DOI: 10.1038/srep12520] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
Atypical Protein Kinase C zeta (PKCζ) forms Partitioning-defective (PAR) polarity complex for apico-basal distribution of membrane proteins essential to maintain normal cellular junctional complexes and tissue homeostasis. Consistently, tumor suppressive role of PKCζ has been established for multiple human cancers. However, recent studies also indicate pro-oncogenic function of PKCζ without firm understanding of detailed molecular mechanism. Here we report a possible mechanism of oncogenic PKCζ signaling in the context of breast cancer. We observed that depletion of PKCζ promotes epithelial morphology in mesenchymal-like MDA-MB-231 cells. The induction of epithelial morphology is associated with significant upregulation of adherens junction (AJ) protein E-cadherin and tight junction (TJ) protein Zonula Occludens-1 (ZO-1). Functionally, depletion of PKCζ significantly inhibits invasion and metastatic progression. Consistently, we observed higher expression and activation of PKCζ signaling in invasive and metastatic breast cancers compared to non-invasive diseases. Mechanistically, an oncogenic PKCζ– NFκB-p65 signaling node might be involved to suppress E-cadherin and ZO-1 expression and ectopic expression of a constitutively active form of NFκB-p65 (S536E-NFκB-p65) significantly rescues invasive potential of PKCζ-depleted breast cancer cells. Thus, our study discovered a PKCζ - NFκB-p65 signaling pathway might be involved to alter cellular junctional dynamics for breast cancer invasive progression.
Collapse
|
37
|
Ma R, Wang C, Wang J, Wang D, Xu J. miRNA-mRNA interaction network in non-small-cell lung cancer. Interdiscip Sci 2015. [PMID: 25863966 DOI: 10.1007/s12539-014-0259-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2014] [Revised: 01/03/2015] [Accepted: 02/09/2015] [Indexed: 11/24/2022]
Abstract
MicroRNAs (miRNA) are small RNA molecules, about 20-25 nucleotides in length. They repress or degrade messenger RNA (mRNA) translation, which are involved in human cancer. In this study based on paired miRNA and mRNA expression profiles of non-small-cell lung cancers (NSCLC) samples, we constructed and analyzed miRNA-mRNA interaction network via several bioinformatics softwares and platforms. This integrative network is comprised of 249 nodes for mRNA, 90 nodes for miRNA and 290 edges that show regulations between target genes and miRNAs. The three miR-1207-5p, miR-1228* and miR-939 are the most connected miRNA that regulated a large number of genes. ST8SIA2, MED1 and HDAC4, SPN, which are targeted by multiple miRNAs and located in the center of the network, are involved in both lung cancer and nervous system via functional annotation analysis. Such a global interaction network of miRNA-mRNA in lung cancer will contribute to refining miRNA target predictions and developing novel therapeutic candidates.
Collapse
Affiliation(s)
- Ruiqi Ma
- College of Bioengineering, Henan University of Technology, Zhengzhou, 450001, China
| | | | | | | | | |
Collapse
|
38
|
The RNA binding protein FXR1 is a new driver in the 3q26-29 amplicon and predicts poor prognosis in human cancers. Proc Natl Acad Sci U S A 2015; 112:3469-74. [PMID: 25733852 DOI: 10.1073/pnas.1421975112] [Citation(s) in RCA: 75] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/27/2023] Open
Abstract
Aberrant expression of RNA-binding proteins has profound implications for cellular physiology and the pathogenesis of human diseases such as cancer. We previously identified the Fragile X-Related 1 gene (FXR1) as one amplified candidate driver gene at 3q26-29 in lung squamous cell carcinoma (SCC). FXR1 is an autosomal paralog of Fragile X mental retardation 1 and has not been directly linked to human cancers. Here we demonstrate that FXR1 is a key regulator of tumor progression and its overexpression is critical for nonsmall cell lung cancer (NSCLC) cell growth in vitro and in vivo. We identified the mechanisms by which FXR1 executes its regulatory function by forming a novel complex with two other oncogenes, protein kinase C, iota and epithelial cell transforming 2, located in the same amplicon via distinct binding mechanisms. FXR1 expression is a candidate biomarker predictive of poor survival in multiple solid tumors including NSCLCs. Because FXR1 is overexpressed and associated with poor clinical outcomes in multiple cancers, these results have implications for other solid malignancies.
Collapse
|
39
|
Guyer RA, Macara IG. Loss of the polarity protein PAR3 activates STAT3 signaling via an atypical protein kinase C (aPKC)/NF-κB/interleukin-6 (IL-6) axis in mouse mammary cells. J Biol Chem 2015; 290:8457-68. [PMID: 25657002 DOI: 10.1074/jbc.m114.621011] [Citation(s) in RCA: 27] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
PAR3 suppresses tumor growth and metastasis in vivo and cell invasion through matrix in vitro. We propose that PAR3 organizes and limits multiple signaling pathways and that inappropriate activation of these pathways occurs without PAR3. Silencing Pard3 in conjunction with oncogenic activation promotes invasion and metastasis via constitutive STAT3 activity in mouse models, but the mechanism for this is unknown. We now show that loss of PAR3 triggers increased production of interleukin-6, which induces STAT3 signaling in an autocrine manner. Activation of atypical protein kinase C ι/λ (aPKCι/λ) mediates this effect by stimulating NF-κB signaling and IL-6 expression. Our results suggest that PAR3 restrains aPKCι/λ activity and thus prevents aPKCι/λ from activating an oncogenic signaling network.
Collapse
Affiliation(s)
- Richard A Guyer
- From the Department of Cell and Developmental Biology and Medical-Scientist Training Program, Vanderbilt University, Nashville, Tennessee 37232
| | - Ian G Macara
- From the Department of Cell and Developmental Biology and
| |
Collapse
|
40
|
Song KB, Liu WJ, Jia SS. miR-219 inhibits the growth and metastasis of TSCC cells by targeting PRKCI. Int J Clin Exp Med 2014; 7:2957-2965. [PMID: 25356169 PMCID: PMC4211819] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2014] [Accepted: 08/28/2014] [Indexed: 06/04/2023]
Abstract
Tongue squamous cells carcinoma (TSCC) is the most common type in oral cancers. Recently, accumulating evidence suggests that microRNAs (miRNAs) play critical roles in tumorigenesis. Here, we demonstrated that miR-219 was significantly downregulated in TSCC tissues and cell lines. miR-219 overexpression remarkably suppressed cell proliferation, colony formation, migration and invasion of TSCC cells. In addition, protein kinase CI (PRKCI) was identified as a target of miR-219, and overexpression of PRKCI could significantly attenuated the tumor suppressive effects of miR-219. Furthermore, PRKCI inversely correlates with miR-219 in TSCC tissues. Taken together, miR-219 inhibited growth and metastasis by targeting PRKCI and might be used as a potential target for the treatment of TSCC.
Collapse
Affiliation(s)
- Kai-Bin Song
- Department of Head and Neck Surgery, Third Affiliated Hospital of Harbin Medical UniversityHarbin 150081, Heilongjiang, China
| | - Wen-Juan Liu
- Department of Neurology, The Second Affiliated Hospital of Harbin Medical UniversityHarbin 150001, Heilongjiang, China
| | - Shen-Shan Jia
- Department of Head and Neck Surgery, Third Affiliated Hospital of Harbin Medical UniversityHarbin 150081, Heilongjiang, China
| |
Collapse
|
41
|
Effects of aurothiomalate treatment on canine osteosarcoma in a murine xenograft model. Anticancer Drugs 2014; 25:332-9. [PMID: 24304691 DOI: 10.1097/cad.0000000000000061] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Osteosarcoma is a highly fatal cancer, with most patients ultimately succumbing to metastatic disease. The purpose of this study was to evaluate the effects of the antirheumatoid drug aurothiomalate on canine and human osteosarcoma cells and on canine osteosarcoma growth and metastasis in a mouse xenograft model. We hypothesized that aurothiomalate would decrease osteosarcoma cell survival, tumor cellular proliferation, tumor growth, and metastasis. After performing clonogenic assays, aurothiomalate or a placebo was administered to 54 mice inoculated with canine osteosarcoma. Survival, tumor growth, embolization, metastasis, histopathology, cell proliferation marker Ki67, and apoptosis marker caspase-3 were compared between groups. Statistical analysis was carried out using the Kaplan-Meier method with the log-rank test and one-way analysis of variance with the Tukey's test or Dunn's method. Aurothiomalate caused dose-dependent inhibition of osteosarcoma cell survival (P<0.001) and decreased tumor growth (P<0.001). Pulmonary macrometastasis and Ki67 labeling were reduced with low-dose aurothiomalate (P=0.033 and 0.005, respectively), and tumor emboli and pulmonary micrometastases were decreased with high-dose aurothiomalate (P=0.010 and 0.011, respectively). There was no difference in survival, tumor development, ulceration, mitotic indices, tumor necrosis, nonpulmonary metastases, and caspase-3 labeling. Aurothiomalate treatment inhibited osteosarcoma cell survival and reduced tumor cell proliferation, growth, embolization, and pulmonary metastasis. Given aurothiomalate's established utility in canine and human medicine, our results suggest that this compound may hold promise as an adjunctive therapy for osteosarcoma. Further translational research is warranted to better characterize the dose response of canine and human osteosarcoma to aurothiomalate.
Collapse
|
42
|
McCray AN, Desai S, Acevedo-Duncan M. The interruption of PKC-ι signaling and TRAIL combination therapy against glioblastoma cells. Neurochem Res 2014; 39:1691-701. [PMID: 24965532 DOI: 10.1007/s11064-014-1361-8] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2013] [Revised: 05/09/2014] [Accepted: 06/12/2014] [Indexed: 12/22/2022]
Abstract
Glioblastoma is a highly aggressive type of brain cancer which currently has limited options for treatment. It is imperative to develop combination therapies that could cause apoptosis in glioblastoma. The aim of this study was to characterize the affect of modified ICA-1, a PKC-iota inhibitor, on the growth pattern of various glioblastoma cell lines. T98G and U87 glioblastoma cells were treated with ICA-1 alone and the absolute cell numbers of each group were determined for cell growth expansion analysis, cell viability analysis, and cell death analysis. Low dose ICA-1 treatment alone significantly inhibited cell growth expansion of high density glioblastoma cells without inducing cell death. However, the high dose ICA-1 treatment regimen provided significant apoptosis for glioblastoma cells. Furthermore, this study was conducted to use a two layer molecular level approach for treating glioblastoma cells with ICA-1 plus an apoptosis agent, tumor-necrosis factor-related apoptosis-inducing ligand (TRAIL), to induce apoptosis in such chemo-refractory cancer cells. Following ICA-1 plus TRAIL treatment, apoptosis was detected in glioblastoma cells via the TUNEL assay and via flow cytometric analysis using Annexin-V FITC/PI. This study offers the first evidence for ICA-1 alone to inhibit glioblastoma cell proliferation as well as the novel combination of ICA-1 with TRAIL to cause robust apoptosis in a caspase-3 mediated mechanism. Furthermore, ICA-1 plus TRAIL simultaneously modulates down-regulation of PKC-iota and c-Jun.
Collapse
Affiliation(s)
- Andrea N McCray
- James A. Haley Veterans' Hospital, 13000 Bruce B. Downs Blvd., VAR 151, Tampa, FL, 33612, USA,
| | | | | |
Collapse
|
43
|
Gont A, Hanson JEL, Lavictoire SJ, Parolin DA, Daneshmand M, Restall IJ, Soucie M, Nicholas G, Woulfe J, Kassam A, Da Silva VF, Lorimer IAJ. PTEN loss represses glioblastoma tumor initiating cell differentiation via inactivation of Lgl1. Oncotarget 2014; 4:1266-79. [PMID: 23907540 PMCID: PMC3787156 DOI: 10.18632/oncotarget.1164] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022] Open
Abstract
Glioblastoma multiforme is an aggressive and incurable type of brain tumor. A subset of undifferentiated glioblastoma cells, known as glioblastoma tumor initiating cells (GTICs), has an essential role in the malignancy of this disease and also appears to mediate resistance to radiation therapy and chemotherapy. GTICs retain the ability to differentiate into cells with reduced malignant potential, but the signaling pathways controlling differentiation are not fully understood at this time. PTEN loss is a very common in glioblastoma multiforme and leads to aberrant activation of the phosphoinositide 3-kinase pathway. Increased signalling through this pathway leads to activation of multiple protein kinases, including atypical protein kinase C. In Drosophila, active atypical protein kinase C has been shown to promote the self-renewal of neuroblasts, inhibiting their differentiation along a neuronal lineage. This effect is mediated by atypical protein kinase c-mediated phosphorylation and inactivation of Lgl, a protein that was first characterized as a tumour suppressor in Drosophila. The effects of the atypical protein kinase C/Lgl pathway on the differentiation status of GTICs, and its potential link to PTEN loss, have not been assessed previously. Here we show that PTEN loss leads to the phosphorylation and inactivation of Lgl by atypical protein kinase C in glioblastoma cells. Re-expression of PTEN in GTICs promoted their differentiation along a neuronal lineage. This effect was also seen when atypical protein kinase C was knocked down using RNA interference, and when a non-phosphorylatable, constitutively active form of Lgl was expressed in GTICs. Thus PTEN loss, acting via atypical protein kinase C activation and Lgl inactivation, helps to maintain GTICs in an undifferentiated state.
Collapse
Affiliation(s)
- Alexander Gont
- Centre for Cancer Therapeutics, Ottawa Hospital Research Institute, 501 Smyth Road, Ottawa, K1H 8L6, Canada
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Tisdale EJ, Shisheva A, Artalejo CR. Overexpression of atypical protein kinase C in HeLa cells facilitates macropinocytosis via Src activation. Cell Signal 2014; 26:1235-42. [PMID: 24582589 PMCID: PMC4149413 DOI: 10.1016/j.cellsig.2014.02.014] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2014] [Accepted: 02/18/2014] [Indexed: 01/08/2023]
Abstract
Atypical protein kinase C (aPKC) is the first recognized kinase oncogene. However, the specific contribution of aPKC to cancer progression is unclear. The pseudosubstrate domain of aPKC is different from the other PKC family members, and therefore a synthetic peptide corresponding to the aPKC pseudosubstrate (aPKC-PS) sequence, which specifically blocks aPKC kinase activity, is a valuable tool to assess the role of aPKC in various cellular processes. Here, we learned that HeLa cells incubated with membrane permeable aPKC-PS peptide displayed dilated heterogeneous vesicles labeled with peptide that were subsequently identified as macropinosomes. A quantitative membrane binding assay revealed that aPKC-PS peptide stimulated aPKC recruitment to membranes and activated Src. Similarly, aPKC overexpression in transfected HeLa cells activated Src and induced macropinosome formation. Src-aPKC interaction was essential; substitution of the proline residues in aPKC that associate with the Src-SH3 binding domain rendered the mutant kinase unable to induce macropinocytosis in transfected cells. We propose that aPKC overexpression is a contributing factor to cell transformation by interacting with and consequently promoting Src activation and constitutive macropinocytosis, which increases uptake of extracellular factors, required for altered cell growth and accelerated cell migration.
Collapse
Affiliation(s)
- Ellen J Tisdale
- Department of Pharmacology, Wayne State University School of Medicine, 540 E. Canfield Ave., 6374 Scott Hall, Detroit, MI 48201, USA.
| | - Assia Shisheva
- Department of Physiology, Wayne State University School of Medicine, 540 E. Canfield Ave., 5374 Scott Hall, Detroit, MI 48201, USA
| | - Cristina R Artalejo
- Department of Pharmacology, Wayne State University School of Medicine, 540 E. Canfield Ave., 6374 Scott Hall, Detroit, MI 48201, USA
| |
Collapse
|
45
|
Pu Y, Kang JH, Sigano DM, Peach M, Lewin NE, Marquez VE, Blumberg PM. Diacylglycerol lactones targeting the structural features that distinguish the atypical C1 domains of protein kinase C ζ and ι from typical C1 domains. J Med Chem 2014; 57:3835-44. [PMID: 24684293 PMCID: PMC4310642 DOI: 10.1021/jm500165n] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2014] [Indexed: 01/25/2023]
Abstract
To explore the feasibility of developing ligands targeted to the atypical C1 domains of protein kinase C ζ and ι, we have prepared diacylglycerol lactones substituted with hydrophilic groups on their side chains, which potentially could interact with the arginine residues that distinguish the atypical C1 domains of PKCζ and PKCι from typical C1 domains, and we have measured their binding to mutated versions of the C1b domain of PKCδ that incorporate one or more of these arginine residues. The most selective of the diacylglycerol lactones showed only a 10-fold reduction in binding affinity with the triple arginine mutant (N7R/S10R/L20R) compared to the wild-type, whereas phorbol 12,13-dibutyrate showed a 6000-fold loss of affinity. Molecular modeling confirms that these ligands are indeed able to interact with the arginine residues. Our results show that dramatic changes in selectivity can be obtained through appropriate substitution of diacylglycerol lactones.
Collapse
Affiliation(s)
- Yongmei Pu
- Laboratory
of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of
Health, Bethesda, Maryland 20892, United
States
| | - Ji-Hye Kang
- Chemical
Biology Laboratory, Center for Cancer Research, National Cancer Institute at Frederick, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Dina M. Sigano
- Chemical
Biology Laboratory, Center for Cancer Research, National Cancer Institute at Frederick, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Megan
L. Peach
- Chemical
Biology Laboratory, Basic Science Program, Frederick National Laboratory
for Cancer Research, Leidos Biomedical,
Inc., Frederick, Maryland 21702, United
States
| | - Nancy E. Lewin
- Laboratory
of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of
Health, Bethesda, Maryland 20892, United
States
| | - Victor E. Marquez
- Chemical
Biology Laboratory, Center for Cancer Research, National Cancer Institute at Frederick, National Institutes of Health, Frederick, Maryland 21702, United States
| | - Peter M. Blumberg
- Laboratory
of Cancer Biology and Genetics, Center for Cancer Research, National Cancer Institute, National Institutes of
Health, Bethesda, Maryland 20892, United
States
| |
Collapse
|
46
|
Paul A, Gunewardena S, Stecklein SR, Saha B, Parelkar N, Danley M, Rajendran G, Home P, Ray S, Jokar I, Vielhauer GA, Jensen RA, Tawfik O, Paul S. PKCλ/ι signaling promotes triple-negative breast cancer growth and metastasis. Cell Death Differ 2014; 21:1469-81. [PMID: 24786829 DOI: 10.1038/cdd.2014.62] [Citation(s) in RCA: 31] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2013] [Revised: 03/28/2014] [Accepted: 03/31/2014] [Indexed: 12/29/2022] Open
Abstract
Triple-negative breast cancer (TNBC) is a distinct breast cancer subtype defined by the absence of estrogen receptor (ER), progesterone receptor (PR) and epidermal growth factor receptor 2 (HER2/neu), and the patients with TNBC are often diagnosed with higher rates of recurrence and metastasis. Because of the absence of ER, PR and HER2/neu expressions, TNBC patients are insensitive to HER2-directed and endocrine therapies available for breast cancer treatment. Here, we report that expression of atypical protein kinase C isoform, PKCλ/ι, significantly increased and activated in all invasive breast cancer (invasive ductal carcinoma or IDC) subtypes including the TNBC subtype. Because of the lack of targeted therapies for TNBC, we choose to study PKCλ/ι signaling as a potential therapeutic target for TNBC. Our observations indicated that PKCλ/ι signaling is highly active during breast cancer invasive progression, and metastatic breast cancers, the advanced stages of breast cancer disease that developed more frequently in TNBC patients, are also characterized with high levels of PKCλ/ι expression and activation. Functional analysis in experimental mouse models revealed that depletion of PKCλ/ι significantly reduces TNBC growth as well as lung metastatic colonization. Furthermore, we have identified a PKCλ/ι-regulated gene signature consisting of 110 genes, which are significantly associated with indolent to invasive progression of human breast cancer and poor prognosis. Mechanistically, cytokines such as TGFβ and IL1β could activate PKCλ/ι signaling in TNBC cells and depletion of PKCλ/ι impairs NF-κB p65 (RelA) nuclear localization. We observed that cytokine-PKCλ/ι-RelA signaling axis, at least in part, involved in modulating gene expression to regulate invasion of TNBC cells. Overall, our results indicate that induction and activation of PKCλ/ι promote TNBC growth, invasion and metastasis. Thus, targeting PKCλ/ι signaling could be a therapeutic option for breast cancer, including the TNBC subtype.
Collapse
Affiliation(s)
- A Paul
- 1] The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA [2] Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - S Gunewardena
- Department of Physiology, University of Kansas Medical Center, Kansas City, KS, USA
| | - S R Stecklein
- 1] The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA [2] Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - B Saha
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - N Parelkar
- The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - M Danley
- 1] The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA [2] Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - G Rajendran
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - P Home
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - S Ray
- Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - I Jokar
- The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA
| | - G A Vielhauer
- Department of Urology, University of Kansas Medical Center, Kansas City, KS, USA
| | - R A Jensen
- 1] The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA [2] Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - O Tawfik
- 1] The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA [2] Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| | - S Paul
- 1] The University of Kansas Cancer Center, University of Kansas Medical Center, Kansas City, KS, USA [2] Department of Pathology and Laboratory Medicine, University of Kansas Medical Center, Kansas City, KS, USA
| |
Collapse
|
47
|
Tang GW, Altman RB. Knowledge-based fragment binding prediction. PLoS Comput Biol 2014; 10:e1003589. [PMID: 24762971 PMCID: PMC3998881 DOI: 10.1371/journal.pcbi.1003589] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 03/11/2014] [Indexed: 11/18/2022] Open
Abstract
Target-based drug discovery must assess many drug-like compounds for potential activity. Focusing on low-molecular-weight compounds (fragments) can dramatically reduce the chemical search space. However, approaches for determining protein-fragment interactions have limitations. Experimental assays are time-consuming, expensive, and not always applicable. At the same time, computational approaches using physics-based methods have limited accuracy. With increasing high-resolution structural data for protein-ligand complexes, there is now an opportunity for data-driven approaches to fragment binding prediction. We present FragFEATURE, a machine learning approach to predict small molecule fragments preferred by a target protein structure. We first create a knowledge base of protein structural environments annotated with the small molecule substructures they bind. These substructures have low-molecular weight and serve as a proxy for fragments. FragFEATURE then compares the structural environments within a target protein to those in the knowledge base to retrieve statistically preferred fragments. It merges information across diverse ligands with shared substructures to generate predictions. Our results demonstrate FragFEATURE's ability to rediscover fragments corresponding to the ligand bound with 74% precision and 82% recall on average. For many protein targets, it identifies high scoring fragments that are substructures of known inhibitors. FragFEATURE thus predicts fragments that can serve as inputs to fragment-based drug design or serve as refinement criteria for creating target-specific compound libraries for experimental or computational screening.
Collapse
Affiliation(s)
- Grace W. Tang
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
| | - Russ B. Altman
- Department of Bioengineering, Stanford University, Stanford, California, United States of America
- Department of Genetics, Stanford University, Stanford, California, United States of America
- * E-mail:
| |
Collapse
|
48
|
Structural and biochemical insights into the homotypic PB1-PB1 complex between PKCζ and p62. SCIENCE CHINA-LIFE SCIENCES 2013; 57:69-80. [PMID: 24369353 DOI: 10.1007/s11427-013-4592-z] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/17/2013] [Accepted: 11/22/2013] [Indexed: 01/10/2023]
Abstract
The atypical PKC isoforms (ζ and ı) play essential roles in regulating various cellular processes. Both the hetero-interaction between PKCζ and p62 through their N-terminal PB1 domains and the homo-oligomerization of p62 via its PB1 domain are critical for the activation of NF-κB signaling; however, the molecular mechanisms concerning the formation and regulation of these homotypic complexes remain unclear. Here we determined the crystal structure of PKCζ-PB1 in complex with a monomeric p62-PB1 mutant, where the massive electrostatic interactions between the acidic OPCA motif of PKCζ-PB1 and the basic surface of p62-PB1, as well as additional hydrogen bonds, ensure the formation of a stable and specific complex. The PKCζ-p62 interaction is interfered with the modification of a specific Cys of PKCζ by the antiarthritis drug aurothiomalate, though all four cysteine residues in the PKCζ-PB1 domain can be modified in in vitro assay. In addition, detailed structural and biochemical analyses demonstrate that the PB1 domains of aPKCs belong to the type I group, which can depolymerize the high-molecular-weight p62 aggregates into homo-oligomers of lower order. These data together unravel the molecular mechanisms of the homo-or hetero-interactions between p62 and PKCζ and provide the basis for designing inhibitors of NF-κB signaling.
Collapse
|
49
|
Bao B, Ahmad A, Azmi AS, Ali S, Sarkar FH. Overview of cancer stem cells (CSCs) and mechanisms of their regulation: implications for cancer therapy. ACTA ACUST UNITED AC 2013; Chapter 14:Unit 14.25. [PMID: 23744710 DOI: 10.1002/0471141755.ph1425s61] [Citation(s) in RCA: 174] [Impact Index Per Article: 15.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
The identification of small subpopulations of cancer stem cells (CSCs) from blood mononuclear cells in human acute myeloid leukemia (AML) in 1997 was a landmark observation that recognized the potential role of CSCs in tumor aggressiveness. Two critical properties contribute to the functional role of CSCs in the establishment and recurrence of cancerous tumors: their capacity for self-renewal and their potential to differentiate into unlimited heterogeneous populations of cancer cells. These findings suggest that CSCs may represent novel therapeutic targets for the treatment and/or prevention of tumor progression, since they appear to be involved in cell migration, invasion, metastasis, and treatment resistance-all of which lead to poor clinical outcomes. The identification of CSC-specific markers, the isolation and characterization of CSCs from malignant tissues, and targeting strategies for the destruction of CSCs provide a novel opportunity for cancer research. This overview describes the potential implications of several common CSC markers in the identification of CSC subpopulations that are restricted to common malignant diseases, e.g., leukemia, and breast, prostate, pancreatic, and lung cancers. The role of microRNAs (miRNAs) in the regulation of CSC function is also discussed, as are several methods commonly used in CSC research. The potential role of the antidiabetic drug metformin- which has been shown to have effects on CSCs, and is known to function as an antitumor agent-is discussed as an example of this new class of chemotherapeutics.
Collapse
Affiliation(s)
- Bin Bao
- Department of Pathology, Karmanos Cancer Institute, Wayne State University, Detroit, MI, USA
| | | | | | | | | |
Collapse
|
50
|
Leppänen T, Tuominen RK, Moilanen E. Protein kinase C and its inhibitors in the regulation of inflammation: inducible nitric oxide synthase as an example. Basic Clin Pharmacol Toxicol 2013; 114:37-43. [PMID: 24107256 DOI: 10.1111/bcpt.12139] [Citation(s) in RCA: 33] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2013] [Accepted: 09/13/2013] [Indexed: 01/19/2023]
Abstract
Protein kinase C (PKC) is a family of ten isoenzymes that play a crucial role in cellular signal transduction. Studies with PKC knockout animals have revealed that many of the isoenzymes are involved in cell growth, proliferation and differentiation. Several PKC isoenzymes have also been shown to be important mediators in inflammation and immunity, particularly in lymphocyte responses. However, less is known about the role of PKC in the regulation of the expression of inflammatory genes. In inflammatory processes, nitric oxide is primarily produced by inducible nitric oxide synthase (iNOS) in inflammatory cells, such as macrophages. In innate immunity, nitric oxide functions as an effector molecule towards the infectious organisms. Increased levels of nitric oxide are also produced by inflammatory and tissue cells in inflammatory diseases, such as asthma and arthritis. In this MiniReview, the role of PKC isoenzymes in the pathogenesis and as a potential drug target in inflammation will be discussed presenting iNOS as an example of an inflammatory gene regulated by the pleiotropic PKC signalling pathway.
Collapse
Affiliation(s)
- Tiina Leppänen
- The Immunopharmacology Research Group, University of Tampere School of Medicine and Tampere University Hospital, Tampere, Finland
| | | | | |
Collapse
|