1
|
Singh RK, Kumar S, Kumar S, Shukla A, Kumar N, Patel AK, Yadav LK, Kaushalendra, Antiwal M, Acharya A. Potential implications of protein kinase Cα in pathophysiological conditions and therapeutic interventions. Life Sci 2023; 330:121999. [PMID: 37536614 DOI: 10.1016/j.lfs.2023.121999] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2023] [Revised: 07/31/2023] [Accepted: 07/31/2023] [Indexed: 08/05/2023]
Abstract
PKCα is a molecule with many functions that play an important role in cell survival and death to maintain cellular homeostasis. Alteration in the normal functioning of PKCα is responsible for the complicated etiology of many pathologies, including cancer, cardiovascular diseases, kidney complications, neurodegenerative diseases, diabetics, and many others. Several studies have been carried out over the years on this kinase's function, and regulation in normal physiology and pathological conditions. A lot of data with antithetical results have therefore accumulated over time to create a complex framework of physiological implications connected to the PKCα function that needs comprehensive elucidation. In light of this information, we critically analyze the multiple roles played by PKCα in basic cellular processes and their molecular mechanism during various pathological conditions. This review further discusses the current approaches to manipulating PKCα signaling amplitude in the patient's favour and proposed PKCα as a therapeutic target to reverse pathological states.
Collapse
Affiliation(s)
- Rishi Kant Singh
- Lab of Hematopoiesis and Leukemia, KSBS, Indian Institute of Technology, Delhi, New Delhi 110016, India; Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Sanjay Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Sandeep Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Alok Shukla
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Naveen Kumar
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Anand Kumar Patel
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Lokesh Kumar Yadav
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India
| | - Kaushalendra
- Department of Zoology, Pachhunga University College Campus, Mizoram University, Aizawl 796001, India
| | - Meera Antiwal
- Institute of Medical Sciences, Banaras Hindu University, Varanasi 221005, India
| | - Arbind Acharya
- Cancer Immunology Lab, Department of Zoology, Banaras Hindu University, Varanasi 221005, India.
| |
Collapse
|
2
|
Ma SC, Zhang JQ, Yan TH, Miao MX, Cao YM, Cao YB, Zhang LC, Li L. Novel strategies to reverse chemoresistance in colorectal cancer. Cancer Med 2023. [PMID: 36645225 DOI: 10.1002/cam4.5594] [Citation(s) in RCA: 16] [Impact Index Per Article: 16.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2022] [Revised: 12/02/2022] [Accepted: 12/21/2022] [Indexed: 01/17/2023] Open
Abstract
Colorectal cancer (CRC) is a common gastrointestinal malignancy with high morbidity and fatality. Chemotherapy, as traditional therapy for CRC, has exerted well antitumor effect and greatly improved the survival of CRC patients. Nevertheless, chemoresistance is one of the major problems during chemotherapy for CRC and significantly limits the efficacy of the treatment and influences the prognosis of patients. To overcome chemoresistance in CRC, many strategies are being investigated. Here, we review the common and novel measures to combat the resistance, including drug repurposing (nonsteroidal anti-inflammatory drugs, metformin, dichloroacetate, enalapril, ivermectin, bazedoxifene, melatonin, and S-adenosylmethionine), gene therapy (ribozymes, RNAi, CRISPR/Cas9, epigenetic therapy, antisense oligonucleotides, and noncoding RNAs), protein inhibitor (EFGR inhibitor, S1PR2 inhibitor, and DNA methyltransferase inhibitor), natural herbal compounds (polyphenols, terpenoids, quinones, alkaloids, and sterols), new drug delivery system (nanocarriers, liposomes, exosomes, and hydrogels), and combination therapy. These common or novel strategies for the reversal of chemoresistance promise to improve the treatment of CRC.
Collapse
Affiliation(s)
- Shu-Chang Ma
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China.,Department of Physiology and Pharmacology, China Pharmaceutic University, Nanjing, China
| | - Jia-Qi Zhang
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Tian-Hua Yan
- Department of Physiology and Pharmacology, China Pharmaceutic University, Nanjing, China
| | - Ming-Xing Miao
- Department of Physiology and Pharmacology, China Pharmaceutic University, Nanjing, China
| | - Ye-Min Cao
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Yong-Bing Cao
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| | - Li-Chao Zhang
- Department of Pharmacy, Shanghai Municipal Hospital of Traditional Chinese Medicine, Shanghai, China
| | - Ling Li
- Institute of Vascular Disease, Shanghai TCM-Integrated Hospital, Shanghai University of Traditional Chinese Medicine, Shanghai, China
| |
Collapse
|
3
|
Kawano T, Inokuchi J, Eto M, Murata M, Kang JH. Protein Kinase C (PKC) Isozymes as Diagnostic and Prognostic Biomarkers and Therapeutic Targets for Cancer. Cancers (Basel) 2022; 14:5425. [PMID: 36358843 PMCID: PMC9658272 DOI: 10.3390/cancers14215425] [Citation(s) in RCA: 11] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2022] [Revised: 11/02/2022] [Accepted: 11/02/2022] [Indexed: 08/05/2023] Open
Abstract
Protein kinase C (PKC) is a large family of calcium- and phospholipid-dependent serine/threonine kinases that consists of at least 11 isozymes. Based on their structural characteristics and mode of activation, the PKC family is classified into three subfamilies: conventional or classic (cPKCs; α, βI, βII, and γ), novel or non-classic (nPKCs; δ, ε, η, and θ), and atypical (aPKCs; ζ, ι, and λ) (PKCλ is the mouse homolog of PKCι) PKC isozymes. PKC isozymes play important roles in proliferation, differentiation, survival, migration, invasion, apoptosis, and anticancer drug resistance in cancer cells. Several studies have shown a positive relationship between PKC isozymes and poor disease-free survival, poor survival following anticancer drug treatment, and increased recurrence. Furthermore, a higher level of PKC activation has been reported in cancer tissues compared to that in normal tissues. These data suggest that PKC isozymes represent potential diagnostic and prognostic biomarkers and therapeutic targets for cancer. This review summarizes the current knowledge and discusses the potential of PKC isozymes as biomarkers in the diagnosis, prognosis, and treatment of cancers.
Collapse
Affiliation(s)
- Takahito Kawano
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Junichi Inokuchi
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masatoshi Eto
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
- Department of Urology, Graduate School of Medical Sciences, Kyushu University, Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Masaharu Murata
- Center for Advanced Medical Innovation, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | - Jeong-Hun Kang
- Division of Biopharmaceutics and Pharmacokinetics, National Cerebral and Cardiovascular Center Research Institute, 6-1 Shinmachi, Kishibe, Suita, Osaka 564-8565, Japan
| |
Collapse
|
4
|
Identification of Novel Anthracycline Resistance Genes and Their Inhibitors. Pharmaceuticals (Basel) 2021; 14:ph14101051. [PMID: 34681275 PMCID: PMC8540045 DOI: 10.3390/ph14101051] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2021] [Revised: 10/05/2021] [Accepted: 10/11/2021] [Indexed: 11/21/2022] Open
Abstract
Differentially expressed genes have been previously identified by us in multidrug-resistant tumor cells mainly resistant to doxorubicin. In the present study, we exemplarily focused on some of these genes to investigate their causative relationship with drug resistance. HMOX1, NEIL2, and PRKCA were overexpressed by lentiviral-plasmid-based transfection of HEK293 cells. An in silico drug repurposing approach was applied using virtual screening and molecular docking of FDA-approved drugs to identify inhibitors of these new drug-resistant genes. Overexpression of the selected genes conferred resistance to doxorubicin and daunorubicin but not to vincristine, docetaxel, and cisplatin, indicating the involvement of these genes in resistance to anthracyclines but not to a broader MDR phenotype. Using virtual drug screening and molecular docking analyses, we identified FDA-approved compounds (conivaptan, bexarotene, and desloratadine) that were interacting with HMOX1 and PRKCA at even stronger binding affinities than 1-(adamantan-1-yl)-2-(1H-imidazol-1-yl)ethenone and ellagic acid as known inhibitors of HMOX1 and PRKCA, respectively. Conivaptan treatment increased doxorubicin sensitivity of both HMOX1- and PRKCA-transfected cell lines. Bexarotene treatment had a comparable doxorubicin-sensitizing effect in HMOX1-transfected cells and desloratadine in PRKCA-transfected cells. Novel drug resistance mechanisms independent of ABC transporters have been identified that contribute to anthracycline resistance in MDR cells.
Collapse
|
5
|
Shehzad A, Ravinayagam V, AlRumaih H, Aljafary M, Almohazey D, Almofty S, Al-Rashid NA, Al-Suhaimi EA. Application of Three-dimensional (3D) Tumor Cell Culture Systems and Mechanism of Drug Resistance. Curr Pharm Des 2020; 25:3599-3607. [PMID: 31612821 DOI: 10.2174/1381612825666191014163923] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2019] [Accepted: 09/29/2019] [Indexed: 12/12/2022]
Abstract
The in-vitro experimental model for the development of cancer therapeutics has always been challenging. Recently, the scientific revolution has improved cell culturing techniques by applying three dimensional (3D) culture system, which provides a similar physiologically relevant in-vivo model for studying various diseases including cancer. In particular, cancer cells exhibiting in-vivo behavior in a model of 3D cell culture is a more accurate cell culture model to test the effectiveness of anticancer drugs or characterization of cancer cells in comparison with two dimensional (2D) monolayer. This study underpins various factors that cause resistance to anticancer drugs in forms of spheroids in 3D in-vitro cell culture and also outlines key challenges and possible solutions for the future development of these systems.
Collapse
Affiliation(s)
- Adeeb Shehzad
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Vijaya Ravinayagam
- Scientific Research & Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Hamad AlRumaih
- College of Dentistry, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Meneerah Aljafary
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Dana Almohazey
- Stem Cell Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Sarah Almofty
- Stem Cell Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Noor A Al-Rashid
- Department of Clinical Pharmacy, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| | - Ebtesam A Al-Suhaimi
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.,Stem Cell Research Department, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia
| |
Collapse
|
6
|
Cheng Y, Zhu Y, Xu W, Xu J, Yang M, Chen P, Zhao J, Geng L, Gong S. PKCα in colon cancer cells promotes M1 macrophage polarization via MKK3/6-P38 MAPK pathway. Mol Carcinog 2018; 57:1017-1029. [PMID: 29637628 DOI: 10.1002/mc.22822] [Citation(s) in RCA: 23] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2017] [Revised: 03/17/2018] [Accepted: 04/04/2018] [Indexed: 01/19/2023]
Abstract
Tumor associated macrophages are potential targets of the immune therapy for patients with colon cancer. PKCα acts as a tumor suppressor in the intestine. However, the correlation between PKCα expressed in colon cancer cells and tumor associated macrophages polarization has never been detected. In the present study, the correlation between PKCα expression and level of M1 macrophages was evaluated in human colon cancer tissues. A xenograft mouse model of colon cancer cells with different PKCα expression level was constructed to evaluate the effect of PKCα on M1 macrophages polarization in vivo. Co-culture of colon cancer cells and differentiated macrophages was used to detect the potential interplay in vitro. PKCα regulated production of cytokines which correlated with macrophage polarization and the underlying mechanism was further explored. Our study showed that high PKCα expression in human colon cancer tissues correlated with better prognosis and high M1 macrophage content. PKCα expressed in colon cancer cells inhibited the growth of colon cancer in mice model. PKCα induced macrophages polarized to the M1-like phenotype both in vitro and in vivo. Mechanistically, PKCα targeted P38 via MKK3/6 to promote IL12 and GM-CSF expression which further enhanced M1-like macrophages polarization. In conclusion, this study provided evidence for the first time that PKCα in colon cancer cells play an anticancer action by inducing the polarization of tumor associated macrophages to M1-like phenotype in the tumor microenvironment. PKCα promoted IL12/GM-CSF-mediated M1 polarization through MKK3/6-P38 signaling pathway. Our investigation suggested that modulation of the PKCα signaling pathway might serve as a novel strategy for colon cancer therapy.
Collapse
Affiliation(s)
- Yang Cheng
- Department of Digestive, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Yun Zhu
- Liver Tumor Center, Department of Infectious Diseases and Hepatology Unit, Nanfang Hospital, Southern Medical University, Guangzhou, Guangdong, China
| | - Wanfu Xu
- Department of Digestive, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Jiajia Xu
- Department of Digestive, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China.,Guangzhou Institute of Pediatrics, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Min Yang
- Department of Digestive, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Peiyu Chen
- Department of Digestive, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Junhong Zhao
- Department of Digestive, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Lanlan Geng
- Department of Digestive, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| | - Sitang Gong
- Department of Digestive, Guangzhou Women and Children's Medical Center, Guangzhou Medical University, Guangzhou, Guangdong, China
| |
Collapse
|
7
|
Lin J, Zhao C, Liu C, Fu S, Han L, Lu X, Yang C. Redox-responsive F127-folate/F127-disulfide bond-d-α-tocopheryl polyethylene glycol 1000 succinate/P123 mixed micelles loaded with paclitaxel for the reversal of multidrug resistance in tumors. Int J Nanomedicine 2018; 13:805-830. [PMID: 29445276 PMCID: PMC5808690 DOI: 10.2147/ijn.s152395] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023] Open
Abstract
Introduction The development of nanodrug carriers utilizing tumor microenvironment has become a hotspot in reversing multidrug resistance (MDR). Materials and methods This study synthesized a redox-sensitive copolymer, Pluronic F127-disulfide bond-d-α-tocopheryl polyethylene glycol 1000 succinate (FSST), through the connection of the reduction-sensitive disulfide bond between F127 and d-α-tocopheryl polyethylene glycol 1000 succinate. This polymer could induce the elevation of reactive oxygen species (ROS) levels, ultimately resulting in cytotoxicity. Moreover, the redox-responsive mixed micelles, F127-folate (FA)/FSST/P123 (FFSSTP), based on FSST, Pluronic F127-FA, and Pluronic P123, were prepared to load paclitaxel (PTX). Results The in vitro release study demonstrated that FFSSTP/PTX accelerated the PTX release through the breakage of disulfide bond in reductive environment. In cellular experiment, FFSSTP/PTX induced significant apoptosis in PTX-resistant MCF-7/PTX cells through inhibiting adenosine triphosphate (ATP)-binding cassette proteins from pumping out PTX by interfering with the mitochondrial function and ATP synthesis. Furthermore, FFSSTP/PTX induced apoptosis through elevating the intracellular levels of ROS. Conclusion FFSSTP could become a potential carrier for the treatment of MDR tumors.
Collapse
Affiliation(s)
- Jing Lin
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Chaoyue Zhao
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Cuijuan Liu
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Shiyao Fu
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Luying Han
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Xinping Lu
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang, China
| | - Chunrong Yang
- College of Pharmacy, Jiamusi University, Jiamusi, Heilongjiang, China
| |
Collapse
|
8
|
Zhu J, Hu M, Qiu L. Drug resistance reversal by combretastatin-A4 phosphate loaded with doxorubicin in polymersomes independent of angiogenesis effect. ACTA ACUST UNITED AC 2017; 69:844-855. [PMID: 28425588 DOI: 10.1111/jphp.12725] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2016] [Accepted: 03/12/2017] [Indexed: 12/01/2022]
Abstract
OBJECTIVES This study aimed to evaluate that the polymersomes (Ps-DOX-CA4P) dual-loaded with combretastatin-A4 phosphate (CA4P) and doxorubicin (DOX) overcame drug resistance and sensitized tumour cells to chemotherapeutic drugs. METHODS Ps-DOX-CA4P were prepared by solvent evaporation method using mPEG-b-PLA as carriers. The potential capability of CA4P to reverse DOX resistance was verified by cytotoxicity test, apoptosis assay and cellular uptake of DOX. The comparison between free drugs and drug-loaded polymersomes was also made on a single-layer cell model and multicellular tumour spheroids to display the superiority of the drug vehicles. Furthermore, we put the emphasis on the investigation into underlying mechanisms for CA4P overcoming DOX resistance. KEY FINDINGS Results showed Ps-DOX-CA4P achieved increased uptake of DOX, enhanced cytotoxicity and apoptotic rate in MCF-7/ADR cells as well as MCF-7/ADR tumour spheroids. The potential molecular mechanisms may be related to inhibiting P-glycoprotein function by downregulating protein kinase Cα, stimulating ATPase activity, depleting ATP and increasing intracellular reactive oxygen species levels. CONCLUSIONS The findings validated the sensitization property of CA4P on DOX independent of its well-known angiogenesis effect, which would provide a novel and promising strategy for drug-resistant cancer therapy.
Collapse
Affiliation(s)
- Jinfang Zhu
- College of Food Science and Pharmaceutical Science, Xinjiang Agricultural University, Urumqi, China.,College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Mengying Hu
- College of Pharmaceutical Sciences, Zhejiang University, Hangzhou, China
| | - Liyan Qiu
- Ministry of Educational (MOE) Key Laboratory of Macromolecular Synthesis and Functionalization, Department of Polymer Science and Engineering, Zhejiang University, Hangzhou, China
| |
Collapse
|
9
|
Protein Kinases C-Mediated Regulations of Drug Transporter Activity, Localization and Expression. Int J Mol Sci 2017; 18:ijms18040764. [PMID: 28375174 PMCID: PMC5412348 DOI: 10.3390/ijms18040764] [Citation(s) in RCA: 31] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2017] [Revised: 03/25/2017] [Accepted: 03/27/2017] [Indexed: 01/05/2023] Open
Abstract
Drug transporters are now recognized as major actors in pharmacokinetics, involved notably in drug–drug interactions and drug adverse effects. Factors that govern their activity, localization and expression are therefore important to consider. In the present review, the implications of protein kinases C (PKCs) in transporter regulations are summarized and discussed. Both solute carrier (SLC) and ATP-binding cassette (ABC) drug transporters can be regulated by PKCs-related signaling pathways. PKCs thus target activity, membrane localization and/or expression level of major influx and efflux drug transporters, in various normal and pathological types of cells and tissues, often in a PKC isoform-specific manner. PKCs are notably implicated in membrane insertion of bile acid transporters in liver and, in this way, are thought to contribute to cholestatic or choleretic effects of endogenous compounds or drugs. The exact clinical relevance of PKCs-related regulation of drug transporters in terms of drug resistance, pharmacokinetics, drug–drug interactions and drug toxicity remains however to be precisely determined. This issue is likely important to consider in the context of the development of new drugs targeting PKCs-mediated signaling pathways, for treating notably cancers, diabetes or psychiatric disorders.
Collapse
|
10
|
Long-Term Alteration of Reactive Oxygen Species Led to Multidrug Resistance in MCF-7 Cells. OXIDATIVE MEDICINE AND CELLULAR LONGEVITY 2016; 2016:7053451. [PMID: 28058088 PMCID: PMC5183793 DOI: 10.1155/2016/7053451] [Citation(s) in RCA: 23] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 08/18/2016] [Revised: 10/25/2016] [Accepted: 11/06/2016] [Indexed: 12/13/2022]
Abstract
Reactive oxygen species (ROS) play an important role in multidrug resistance (MDR). This study aimed to investigate the effects of long-term ROS alteration on MDR in MCF-7 cells and to explore its underlying mechanism. Our study showed both long-term treatments of H2O2 and glutathione (GSH) led to MDR with suppressed iROS levels in MCF-7 cells. Moreover, the MDR cells induced by 0.1 μM H2O2 treatment for 20 weeks (MCF-7/ROS cells) had a higher viability and proliferative ability than the control MCF-7 cells. MCF-7/ROS cells also showed higher activity or content of intracellular antioxidants like glutathione peroxidase (GPx), GSH, superoxide dismutase (SOD), and catalase (CAT). Importantly, MCF-7/ROS cells were characterized by overexpression of MDR-related protein 1 (MRP1) and P-glycoprotein (P-gp), as well as their regulators NF-E2-related factor 2 (Nrf2), hypoxia-inducible factor 1 (HIF-1α), and the activation of PI3K/Akt pathway in upstream. Moreover, several typical MDR mediators, including glutathione S-transferase-π (GST-π) and c-Myc and Protein Kinase Cα (PKCα), were also found to be upregulated in MCF-7/ROS cells. Collectively, our results suggest that ROS may be critical in the generation of MDR, which may provide new insights into understanding of mechanisms of MDR.
Collapse
|
11
|
Kadioglu O, Cao J, Kosyakova N, Mrasek K, Liehr T, Efferth T. Genomic and transcriptomic profiling of resistant CEM/ADR-5000 and sensitive CCRF-CEM leukaemia cells for unravelling the full complexity of multi-factorial multidrug resistance. Sci Rep 2016; 6:36754. [PMID: 27824156 PMCID: PMC5099876 DOI: 10.1038/srep36754] [Citation(s) in RCA: 35] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/20/2016] [Accepted: 10/17/2016] [Indexed: 12/18/2022] Open
Abstract
We systematically characterised multifactorial multidrug resistance (MDR) in CEM/ADR5000 cells, a doxorubicin-resistant sub-line derived from drug-sensitive, parental CCRF-CEM cells developed in vitro. RNA sequencing and network analyses (Ingenuity Pathway Analysis) were performed. Chromosomal aberrations were identified by array-comparative genomic hybridisation (aCGH) and multicolour fluorescence in situ hybridisation (mFISH). Fifteen ATP-binding cassette transporters and numerous new genes were overexpressed in CEM/ADR5000 cells. The basic karyotype in CCRF-CEM cells consisted of 47, XX, der(5)t(5;14) (q35.33;q32.3), del(9) (p14.1), +20. CEM/ADR5000 cells acquired additional aberrations, including X-chromosome loss, 4q and 14q deletion, chromosome 7 inversion, balanced and unbalanced two and three way translocations: t(3;10), der(3)t(3;13), der(5)t(18;5;14), t(10;16), der(18)t(7;18), der(18)t(21;18;5), der(21;21;18;5) and der(22)t(9;22). CCRF-CEM consisted of two and CEM/ADR5000 of five major sub-clones, indicating genetic tumor heterogeneity. Loss of 3q27.1 in CEM/ADR5000 caused down-regulation of ABCC5 and ABCF3 expression, Xq28 loss down-regulated ABCD1 expression. ABCB1, the most well-known MDR gene, was 448-fold up-regulated due to 7q21.12 amplification. In addition to well-known drug resistance genes, numerous novel genes and genomic aberrations were identified. Transcriptomics and genetics in CEM/AD5000 cells unravelled a range of MDR mechanisms, which is much more complex than estimated thus far. This may have important implications for future treatment strategies.
Collapse
Affiliation(s)
- Onat Kadioglu
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Jingming Cao
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| | - Nadezda Kosyakova
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Jena, Germany
| | - Kristin Mrasek
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Jena, Germany
| | - Thomas Liehr
- Jena University Hospital, Friedrich Schiller University, Institute of Human Genetics, Jena, Germany
| | - Thomas Efferth
- Department of Pharmaceutical Biology, Institute of Pharmacy and Biochemistry, Johannes Gutenberg University, Mainz, Germany
| |
Collapse
|
12
|
Hypericin in the Dark: Foe or Ally in Photodynamic Therapy? Cancers (Basel) 2016; 8:cancers8100093. [PMID: 27754424 PMCID: PMC5082383 DOI: 10.3390/cancers8100093] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/30/2016] [Revised: 09/29/2016] [Accepted: 10/04/2016] [Indexed: 12/12/2022] Open
Abstract
Photosensitizers (PSs) in photodynamic therapy (PDT) are, in most cases, administered systemically with preferential accumulation in malignant tissues; however, exposure of non-malignant tissues to PS may also be clinically relevant, when PS molecules affect the pro-apoptotic cascade without illumination. Hypericin (Hyp) as PS and its derivatives have long been studied, regarding their photodynamic and photocytotoxic characteristics. Hyp and its derivatives have displayed light-activated antiproliferative and cytotoxic effects in many tumor cell lines without cytotoxicity in the dark. However, light-independent effects of Hyp have emerged. Contrary to the acclaimed Hyp minimal dark cytotoxicity and preferential accumulation in tumor cells, it was recently been shown that non-malignant and malignant cells uptake Hyp at a similar level. In addition, Hyp has displayed light-independent toxicity and anti-proliferative effects in a wide range of concentrations. There are multiple mechanisms underlying Hyp light-independent effects, and we are still missing many details about them. In this paper, we focus on Hyp light-independent effects at several sub-cellular levels—protein distribution and synthesis, organelle ultrastructure and function, and Hyp light-independent effects regarding reactive oxygen species (ROS). We summarize work from our laboratories and that of others to reveal an intricate network of the Hyp light-independent effects. We propose a schematic model of pro- and anti-apoptotic protein dynamics between cell organelles due to Hyp presence without illumination. Based on our model, Hyp can be explored as an adjuvant therapeutic drug in combination with chemo- or radiation cancer therapy.
Collapse
|
13
|
Maurya AK, Vinayak M. PI-103 attenuates PI3K-AKT signaling and induces apoptosis in murineT-cell lymphoma. Leuk Lymphoma 2016; 58:1153-1161. [PMID: 27658642 DOI: 10.1080/10428194.2016.1225207] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
Abstract
Aberrant activation of PI3K-AKT signaling in many pathological conditions including cancer has attracted much of interest for drug targeting. Various isoforms are known from three classes of PI3K. Targeting selective isoform is advantageous to overcome the global deleterious effects of drug. PI-103 is a specific inhibitor of p110α of class I PI3K. The present study is aimed to analyze anti-carcinogenic activity of PI-103 in Dalton's lymphoma ascite (DLA) cells. Result shows regression in cell proliferation and increased apoptosis in terms of increased Annexin V binding, nuclear fragmentation and active caspase 3 level. It is correlated with attenuation of PI3K-AKT signaling by PI-103 via downregulation of the level of p110α, phospho-p85α, phospho- AKT, and PKCα in DLA cells as well as in H2O2 induced DLA cells. Additionally, ROS accumulation is declined in H2O2 induced DLA cells. Overall result suggests that PI-103 attenuates PI3K-AKT signaling via induction of apoptosis in murine T-cell lymphoma.
Collapse
Affiliation(s)
- Akhilendra Kumar Maurya
- a Biochemistry & Molecular Biology Laboratory, Department of Zoology, Institute of Science , Banaras Hindu University , Varanasi , India
| | - Manjula Vinayak
- a Biochemistry & Molecular Biology Laboratory, Department of Zoology, Institute of Science , Banaras Hindu University , Varanasi , India
| |
Collapse
|
14
|
Chen S, Wang Y, Zhang Y, Wan Y. Low expression of PKCα and high expression of KRAS predict poor prognosis in patients with colorectal cancer. Oncol Lett 2016; 12:1655-1660. [PMID: 27602102 PMCID: PMC4998155 DOI: 10.3892/ol.2016.4845] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/11/2015] [Accepted: 03/08/2016] [Indexed: 01/13/2023] Open
Abstract
The current study aimed to determine the association between protein kinase Cα (PKCα) and Kirsten rat sarcoma viral oncogene homolog (KRAS) expression and the response to folinic acid, 5-fluorouracil and oxaliplatin (FOLFOX regimen) in patients with colorectal cancer (CRC). The protein levels of PKCα and KRAS were analyzed by immunohistochemistry in tissue samples from patients with CRC and in non-cancerous tissues, including 152 cases of colorectal adenocarcinoma, 30 cases of colorectal adenoma and 20 normal colonic mucosa samples. The association between PKCα and KRAS expression and clinicopathological features was analyzed. The rates of positive PKCα protein expression in patients with poorly, moderately and well-differentiated adenocarcinoma were 16.7% (6/36), 40.0% (24/60), and 57.1% (32/56), respectively (P<0.013). The rate of positive KRAS expression in CRC patients was significantly higher than in patients with colon adenoma and normal colon mucosa (P<0.001). Expression levels of KRAS were associated with the degree of differentiation of CRC (P<0.001). Expression of PKCα was negatively correlated with KRAS expression in CRC tissues. The mean progression-free survival (PFS) times in patients with high and low expression of PKCα were 43.9 and 38.8 months, respectively (P<0.001). The mean PFS times were 38.5 and 45.5 months in patients with high and low expression of KRAS, respectively (P=0.001). In conclusion, low PKCα and high KRAS expression predicted relatively poor prognosis in patients with CRC.
Collapse
Affiliation(s)
- Suxian Chen
- Department of Pathology, The Third Affiliated Hospital of Liaoning Medical College, Jinzhou, Liaoning 121002, P.R. China
| | - Yadi Wang
- Department of Oncology, The Third Affiliated Hospital of Liaoning Medical College, Jinzhou, Liaoning 121002, P.R. China
| | - Yun Zhang
- Department of Obstetrics and Gynecology, The Third Affiliated Hospital of Liaoning Medical College, Jinzhou, Liaoning 121002, P.R. China
| | - Yizeng Wan
- Department of Pathology, The Third Affiliated Hospital of Liaoning Medical College, Jinzhou, Liaoning 121002, P.R. China
- Correspondence to: Dr Yizeng Wan, Department of Pathology, The Third Affiliated Hospital of Liaoning Medical College, 2 Heping Road Section 5, Linghe, Jinzhou, Liaoning 121002, P.R. China, E-mail:
| |
Collapse
|
15
|
Verebova V, Belej D, Joniova J, Jurasekova Z, Miskovsky P, Kozar T, Horvath D, Stanicova J, Huntosova V. Deeper insights into the drug defense of glioma cells against hydrophobic molecules. Int J Pharm 2016; 503:56-67. [PMID: 26940808 DOI: 10.1016/j.ijpharm.2016.02.042] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2015] [Revised: 02/24/2016] [Accepted: 02/26/2016] [Indexed: 12/28/2022]
Abstract
By means of fluorescence microscopy the intracellular distribution of fluorescent drugs with different hydrophobicity (quinizarin, emodin and hypericin) was studied. Selective photoactivation of these drugs in precisely defined position (nuclear envelope) allowed moderately hydrophobic emodin enter the nucleus. Highly hydrophobic hypericin was predominantly kept in the membranes with no fluorescence observed in the nucleus. The redistribution of quinizarin, emodin and hypericin between lipids, proteins and DNA was studied in solutions and cells. Based on these results was proposed theoretical model of hydrophobic drugs' nuclear internalization after photo-activation. Molecular docking models showed that hypericin has the strongest affinity to P-glycoprotein involved in the cell detoxification. Presence of 10 μM quinizarin, emodin or hypericin increased P-glycoprotein function in U87 MG cells. Moreover, emodin pretreatment allowed quinizarin nuclear internalization without photo-activation, which was not the case for hypericin. The synergy of such pretreatment and photo-activation should lessen the drug doses with simultaneous increase of drug efficacy triggering cell apoptosis/necrosis.
Collapse
Affiliation(s)
- Valeria Verebova
- Institute of Biophysics, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovakia.
| | - Dominik Belej
- Department of Biophysics, Faculty of Science, P. J. Safarik University in Kosice, Jesenna 5, 041 54 Kosice, Slovakia.
| | - Jaroslava Joniova
- Department of Biophysics, Faculty of Science, P. J. Safarik University in Kosice, Jesenna 5, 041 54 Kosice, Slovakia.
| | - Zuzana Jurasekova
- Department of Biophysics, Faculty of Science, P. J. Safarik University in Kosice, Jesenna 5, 041 54 Kosice, Slovakia; Center for Interdisciplinary Biosciences, Faculty of Science, P. J. Safarik University in Kosice, Jesenna 5, 041 54 Kosice, Slovakia.
| | - Pavol Miskovsky
- Department of Biophysics, Faculty of Science, P. J. Safarik University in Kosice, Jesenna 5, 041 54 Kosice, Slovakia; Center for Interdisciplinary Biosciences, Faculty of Science, P. J. Safarik University in Kosice, Jesenna 5, 041 54 Kosice, Slovakia.
| | - Tibor Kozar
- Center for Interdisciplinary Biosciences, Faculty of Science, P. J. Safarik University in Kosice, Jesenna 5, 041 54 Kosice, Slovakia.
| | - Denis Horvath
- Center for Interdisciplinary Biosciences, Faculty of Science, P. J. Safarik University in Kosice, Jesenna 5, 041 54 Kosice, Slovakia.
| | - Jana Stanicova
- Institute of Biophysics, University of Veterinary Medicine and Pharmacy, Komenskeho 73, 041 81 Kosice, Slovakia; Institute of Biophysics and Informatics, First Faculty of Medicine, Charles University in Prague, Salmovska 1, 120 00 Prague 2, Czech Republic.
| | - Veronika Huntosova
- Center for Interdisciplinary Biosciences, Faculty of Science, P. J. Safarik University in Kosice, Jesenna 5, 041 54 Kosice, Slovakia.
| |
Collapse
|
16
|
Yang M, Jiang D, Chen Z, Chen J. Photodynamic therapy of drug-resistant human colon adenocarcinoma using verteporfin-loaded TPGS nanoparticles with tumor homing and penetrating peptide functionalization. RSC Adv 2016. [DOI: 10.1039/c6ra19152e] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022] Open
Abstract
tLyp-1-modified nanoparticles loaded with VP (t-NP) as a dual-targeting drug delivery systemviaNRP-1-mediated endocytosis and penetration.
Collapse
Affiliation(s)
- Mengshi Yang
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Material Science and Engineering
- Donghua University
- Shanghai
- China
| | - Di Jiang
- Key Laboratory of Smart Drug Delivery
- School of Pharmacy
- Fudan University
- Shanghai 201203
- China
| | - Zhilong Chen
- State Key Laboratory for Modification of Chemical Fibers and Polymer Materials
- College of Material Science and Engineering
- Donghua University
- Shanghai
- China
| | - Jun Chen
- Key Laboratory of Smart Drug Delivery
- School of Pharmacy
- Fudan University
- Shanghai 201203
- China
| |
Collapse
|
17
|
Anticarcinogenic action of quercetin by downregulation of phosphatidylinositol 3-kinase (PI3K) and protein kinase C (PKC) via induction of p53 in hepatocellular carcinoma (HepG2) cell line. Mol Biol Rep 2015; 42:1419-29. [DOI: 10.1007/s11033-015-3921-7] [Citation(s) in RCA: 74] [Impact Index Per Article: 8.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2015] [Accepted: 08/20/2015] [Indexed: 02/06/2023]
|
18
|
TRM6/61 connects PKCα with translational control through tRNAiMet stabilization: impact on tumorigenesis. Oncogene 2015; 35:1785-96. [DOI: 10.1038/onc.2015.244] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2014] [Revised: 04/29/2015] [Accepted: 05/16/2015] [Indexed: 12/17/2022]
|
19
|
Maurya AK, Vinayak M. Modulation of PKC signaling and induction of apoptosis through suppression of reactive oxygen species and tumor necrosis factor receptor 1 (TNFR1): key role of quercetin in cancer prevention. Tumour Biol 2015; 36:8913-24. [DOI: 10.1007/s13277-015-3634-5] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/16/2015] [Accepted: 06/01/2015] [Indexed: 02/06/2023] Open
|
20
|
Abera MB, Kazanietz MG. Protein kinase Cα mediates erlotinib resistance in lung cancer cells. Mol Pharmacol 2015; 87:832-41. [PMID: 25724832 DOI: 10.1124/mol.115.097725] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
Overexpression and mutational activation of the epidermal growth factor receptor (EGFR) plays an important role in the pathogenesis of non-small cell lung cancer (NSCLC). EGFR tyrosine-kinase inhibitors (TKIs) are given as a primary therapy for advanced patients with EGFR-activating mutations; however, the majority of these tumors relapse and patients eventually develop resistance to TKIs. To address a potential role of protein kinase C (PKC) isozymes in the resistance to TKIs, we used the isogenic NSCLC H1650 cell line and its erlotinib-resistant derivative H1650-M3, a cell line that displays a mesenchymal-like morphology driven by transforming growth factor-β signaling. We found that H1650-M3 cells display remarkable PKCα upregulation and PKCδ downregulation. Notably, silencing PKCα from H1650-M3 cells using RNA interference caused a significant reduction in the expression of epithelial-to-mesenchymal transition (EMT) markers vimentin, Zeb2, Snail, and Twist. Moreover, pharmacological inhibition or PKCα RNA interference depletion and PKCδ restoring sensitized H1650-M3 cells to erlotinib. Whereas ectopic overexpression of PKCα in parental H1650 cells was not sufficient to alter the expression of EMT genes or to confer resistance to erlotinib, it caused downregulation of PKCδ expression, suggesting a unidirectional crosstalk. Finally, mechanistic studies revealed that PKCα upregulation in H1650-M3 cells is driven by transforming growth factor-β. Our results identified important roles for specific PKC isozymes in erlotinib resistance and EMT in lung cancer cells, and highlight PKCα as a potential target for lung cancer treatment.
Collapse
Affiliation(s)
- Mahlet B Abera
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| | - Marcelo G Kazanietz
- Department of Systems Pharmacology and Translational Therapeutics, Perelman School of Medicine, University of Pennsylvania, Philadelphia, Pennsylvania
| |
Collapse
|
21
|
Luna–López A, González-Puertos VY, López-Diazguerrero NE, Königsberg M. New considerations on hormetic response against oxidative stress. J Cell Commun Signal 2014; 8:323-31. [PMID: 25284448 PMCID: PMC4390794 DOI: 10.1007/s12079-014-0248-4] [Citation(s) in RCA: 34] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2014] [Accepted: 09/30/2014] [Indexed: 01/06/2023] Open
Abstract
In order to survive living organisms have developed multiple mechanisms to deal with tough environmental conditions. Hormesis is defined as a process in which exposure to a low dose of a chemical agent or environmental factor that is damaging at higher doses induces an adaptive beneficial effect on the cell or organism. In this paper, we examine several ideas that might be taken into consideration before using hormesis as a therapeutic tool to improve health and life span, and hopefully will open the discussion for new and interesting debates regard hormesis. The first one is to understand that the same stressor or inductor can activate different pathways in a parallel or dual response, which might lead to diverse outcomes. Another idea is related to the mechanisms involved in activating Nrf2, which might be different and have diverse hormetic effects.Last, we discuss mild oxidative stress in association to low-grade chronic inflammation as a stimulating avenue to be explored and the unexpected effects proposed by the obesity paradox theory. All the previous might help to clarify the reasons why centenarians are able to reach the extreme limits of human life span, which could probably be related to the way they deal with homeostasis maintenance, providing an opportunity for hormesis to intervene significantly.
Collapse
Affiliation(s)
| | - Viridiana Y. González-Puertos
- />Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, A.P. 55-535, C.P 09340 México, D.F Mexico
| | - Norma E. López-Diazguerrero
- />Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, A.P. 55-535, C.P 09340 México, D.F Mexico
| | - Mina Königsberg
- />Departamento de Ciencias de la Salud, División de Ciencias Biológicas y de la Salud, Universidad Autónoma Metropolitana-Iztapalapa, A.P. 55-535, C.P 09340 México, D.F Mexico
| |
Collapse
|
22
|
The inhibitory effect of pseudolaric acid B on gastric cancer and multidrug resistance via Cox-2/PKC-α/P-gp pathway. PLoS One 2014; 9:e107830. [PMID: 25250794 PMCID: PMC4176716 DOI: 10.1371/journal.pone.0107830] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2014] [Accepted: 08/20/2014] [Indexed: 01/08/2023] Open
Abstract
Aim To investigate the inhibitory effect of pseudolaric acid B on subcutaneous xenografts of human gastric adenocarcinoma and the underlying molecular mechanisms involved in its multidrug resistance. Methods Human gastric adenocarcinoma SGC7901 cells and drug-resistant SGC7901/ADR cells were injected into nude mice to establish a subcutaneous xenograft model. The effects of pseudolaric acid B with or without adriamycin treatment were compared by determining the tumor size and weight. Cyclo-oxygenase-2, protein kinaseC-α and P-glycoprotein expression levels were determined by immunohistochemistry and western blot. Results Pseudolaric acid B significantly suppressed the tumor growth induced by SGC7901 cells and SGC7901/ADR cells. The combination of pseudolaric acid B and the traditional chemotherapy drug adriamycin exhibited more potent inhibitory effects on the growth of gastric cancer in vivo than treatment with either pseudolaric acid B or adriamycin alone. Protein expression levels of cyclo-oxygenase-2, protein kinaseC-α and P-glycoprotein were inhibited by pseudolaric acid B alone or in combination with adriamycin in SGC7901/ADR cell xenografts. Conclusion Pseudolaric acid B has a significant inhibitory effect and an additive inhibitory effect in combination with adriamycin on the growth of gastric cancer in vivo, which reverses the multidrug resistance of gastric neoplasm to chemotherapy drugs by downregulating the Cox-2/PKC-α/P-gp/mdr1 signaling pathway.
Collapse
|
23
|
Hung TH, Chen CM, Tseng CP, Shen CJ, Wang HL, Choo KB, Chong KY. FZD1 activates protein kinase C delta-mediated drug-resistance in multidrug-resistant MES-SA/Dx5 cancer cells. Int J Biochem Cell Biol 2014; 53:55-65. [PMID: 24814288 DOI: 10.1016/j.biocel.2014.04.011] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/10/2013] [Revised: 01/16/2014] [Accepted: 04/10/2014] [Indexed: 12/13/2022]
Abstract
Multidrug-resistant (MDR) cancer is a major clinical problem in chemotherapy of cancer patients. We have noted inappropriate PKCδ hypomethylation and overexpression of genes in the PKCδ/AP-1 pathway in the human uterus sarcoma drug-resistant cell line, MES-SA/Dx5 cells, which also overexpress p-glycoprotein (ABCB1). Recent studies have indicated that FZD1 is overexpressed in both multidrug-resistant cancer cell lines and in clinical tumor samples. These data have led us to hypothesize that the FZD1-mediated PKCδ signal-transduction pathway may play an important role in drug resistance in MES-SA/Dx5 cells. In this work, the PKCδ inhibitor Rottlerin was found to reduce ABCB1 expression and to inhibit the MDR drug pumping ability in the MES-SA/Dx5 cells when compared with the doxorubicin-sensitive parental cell line, MES-SA. PKCδ was up-regulated with concurrent up-regulation of the mRNA levels of the AP-1-related factors, c-JUN and c-FOS. Activation of AP-1 also correlated with up-regulation of the AP-1 downstream genes HGF and EGR1. Furthermore, AP-1 activities were reduced and the AP-1 downstream genes were down-regulated in Rottlerin-treated or PKCδ shRNA-transfected cells. MES-SA/Dx5 cells were resensitized to doxorubicin-induced toxicity by co-treatment with doxorubicin and Rottlerin or PKCδ shRNA. In addition, cell viability and drug pump-out ability were significantly reduced in the FZD1 inhibitor curcumin-treated and FZD1 shRNA-knockdown MES-SA/Dx5 cells, indicating involvement of PKCδ in FZD1-modulated ABCB1 expression pathway. Taken together, our data demonstrate that FZD1 regulates PKCδ, and the PKCδ/AP-1 signalling transduction pathway plays an important role in drug resistance in MES-SA/Dx5 cells.
Collapse
Affiliation(s)
- Tsai-Hsien Hung
- Graduate Institute of Biomedical Sciences, Division of Biotechnology College of medicine, Chang Gung University,Tao-Yuan, Taiwan, Republic of China
| | - Chuan-Mu Chen
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan, Republic of China
| | - Ching-Ping Tseng
- Graduate Institute of Biomedical Sciences, Division of Biotechnology College of medicine, Chang Gung University,Tao-Yuan, Taiwan, Republic of China; Department of Medical Biotechnology and Laboratory Science, College of medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China; Molecular Medicine Research Center, College of medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China
| | - Chih-Jie Shen
- Department of Life Sciences, National Chung Hsing University, Taichung, Taiwan, Republic of China
| | - Hui-Ling Wang
- Department of Medical Biotechnology and Laboratory Science, College of medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China
| | - Kong-Bung Choo
- Department of Preclinical Sciences, Faculty of Medicine and Health Sciences and Centre for Stem Cell Research, Universiti Tunku Abdul Rahman, Selangor, Malaysia
| | - Kowit Yu Chong
- Graduate Institute of Biomedical Sciences, Division of Biotechnology College of medicine, Chang Gung University,Tao-Yuan, Taiwan, Republic of China; Department of Medical Biotechnology and Laboratory Science, College of medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China; Molecular Medicine Research Center, College of medicine, Chang Gung University, Tao-Yuan, Taiwan, Republic of China.
| |
Collapse
|
24
|
Abstract
Protein kinase C (PKC) is a family of phospholipid-dependent serine/threonine kinases, which can be further classified into three PKC isozymes subfamilies: conventional or classic, novel or nonclassic, and atypical. PKC isozymes are known to be involved in cell proliferation, survival, invasion, migration, apoptosis, angiogenesis, and drug resistance. Because of their key roles in cell signaling, PKC isozymes also have the potential to be promising therapeutic targets for several diseases, such as cardiovascular diseases, immune and inflammatory diseases, neurological diseases, metabolic disorders, and multiple types of cancer. This review primarily focuses on the activation, mechanism, and function of PKC isozymes during cancer development and progression.
Collapse
|
25
|
Al-Suhaimi E. Molecular mechanisms of leptin and pro-apoptotic signals induced by menadione in HepG2 cells. Saudi J Biol Sci 2014; 21:582-8. [PMID: 25473367 DOI: 10.1016/j.sjbs.2014.03.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2013] [Revised: 03/19/2014] [Accepted: 03/20/2014] [Indexed: 12/18/2022] Open
Abstract
Apoptosis is a significant physiological function in the cell. P(53) is known as tumor suppressor cellular factor, executive caspases are also the most involved pathway for apoptosis. Menadione (VK3) has apoptotic action on many harmful cells, but the molecular role of adipokines is not studied enough in this regard, so the ability of menadione to modify the adipokine (leptin hormone), caspase-3 and P(53) signals to induce its apoptotic action on HepG2 cells was studied. The study revealed that menadione has anti-viability and apoptotic effect at sub-G1 phase of HepG2 cell cycle. Its cytotoxic effect is mediated by molecular mechanisms included: inhibiting leptin expression and level, activating caspase-3 pathway and up-regulating the expression of P(53). Menadione exerts its apoptotic mechanisms in a concentration and time dependent way through ROS generation. In addition to the known apoptotic pathways, the results indicate that suppressing leptin pathway is a significant mechanism for menadione apoptotic effect which made it as a potential therapeutic vitamin in preventing hepatocyte survival and proliferation.
Collapse
Affiliation(s)
- Ebtesam Al-Suhaimi
- Biology Department, College of Sciences, University of Dammam, Dammam, Saudi Arabia
| |
Collapse
|
26
|
Liu J, Kong CZ, Gong DX, Zhang Z, Zhu YY. PKC α regulates netrin-1/UNC5B-mediated survival pathway in bladder cancer. BMC Cancer 2014; 14:93. [PMID: 24528886 PMCID: PMC3937025 DOI: 10.1186/1471-2407-14-93] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2013] [Accepted: 02/11/2014] [Indexed: 01/23/2023] Open
Abstract
BACKGROUND Netrin-1 and its receptor UNC5B play important roles in angiogenesis, embryonic development, cancer and inflammation. However, their expression patttern and biological roles in bladder cancer have not been well characterized. The present study aims to investigating the clinical significance of PKC α, netrin-1 and UNC5B in bladder cancer as well as their association with malignant biological behavior of cancer cells. METHODS Netrin-1 and UNC5B expression was examined in 120 bladder cancer specimens using immunohistochemistry and in 40 fresh cancer tissues by western blot. Immunofluorescence was performed in cancer cell lines. PKC α agonist PMA and PKC siRNA was employed in bladder cancer cells. CCK-8, wound healing assays and flow cytometry analysis were used to examine cell proliferation, migration and cell cycle, respectively. RESULTS Netrin-1 expression was positively correlated with histological grade, T stage, metastasis and poor prognosis in bladder cancer tissues. Immunofluorescence showed elevated netrin-1 and decreased UNC5B expression in bladder cancer cells compared with normal bladder cell line. Furthermore, cell proliferation, migration and cell cycle progression were promoted with PMA treatment while inhibited by calphostin C. In addition, PMA treatment could induce while calphostin C reduce netrin-1 expression in bladder cancer cells. CONCLUSIONS The present study identified netrin-1/UNC5B, which could be regulated by PKC signaling, was important mediators of bladder cancer progression.
Collapse
Affiliation(s)
| | - Chui-ze Kong
- Department of Urology, the First Affiliated Hospital of China Medical University, Shenyang, Liaoning 110001, China.
| | | | | | | |
Collapse
|
27
|
Shehzad A, Lee J, Huh TL, Lee YS. Curcumin induces apoptosis in human colorectal carcinoma (HCT-15) cells by regulating expression of Prp4 and p53. Mol Cells 2013; 35:526-32. [PMID: 23686430 PMCID: PMC3887881 DOI: 10.1007/s10059-013-0038-5] [Citation(s) in RCA: 60] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/04/2013] [Revised: 03/13/2013] [Accepted: 03/28/2013] [Indexed: 12/30/2022] Open
Abstract
Curcumin (diferuloylmethane), the yellow pigment of turmeric, is one of the most commonly used and extensively studied phytochemicals due to its pleiotropic effects in several human cancers. In the current study, the therapeutic efficacy of curcumin was investigated in human colorectal carcinoma HCT-15 cells. Curcumin inhibited HCT-15 cells proliferation and induced apoptosis in a dose- and time-dependent manner. Hoechst 33342 and DCFHDA staining revealed morphological and biochemical features of apoptosis as well as ROS generation in HCT-15 cells treated with 30 and 50 μM curcumin. Over-expression of pre-mRNA processing factor 4B (Prp4B) and p53 mutations have been reported as hallmarks of cancer cells. Western blot analysis revealed that curcumin treatment activated caspase-3 and decreased expression of p53 and Prp4B in a time-dependent manner. Transfection of HCT-15 cells with Prp4B clone perturbed the growth inhibition induced by 30 μM curcumin. Fractionation of cells revealed increased accumulation of Prp4B in the nucleus, following its translocation from the cytoplasm. To further evaluate the underlying mechanism and survival effect of Prp4B, we generated siRNA-Prp4B HCT15 clones. Knockdown of Prp4B with siRNA diminished the protective effects of Prp4B against curcumin-induced apoptosis. These results suggest a possible underlying molecular mechanism in which Prp4B over-expression and activity are closely associated with the survival and regulation of apoptotic events in human colon cancer HCT-15 cells.
Collapse
Affiliation(s)
- Adeeb Shehzad
- School of Life Sciences, College of Natural Sciences, Kyungpook National University
| | | | - Tae-Lin Huh
- School of Life Sciences, College of Natural Sciences, Kyungpook National University
| | - Young Sup Lee
- School of Life Sciences, College of Natural Sciences, Kyungpook National University
| |
Collapse
|
28
|
Bipiperidinyl derivatives of 23-hydroxybetulinic acid reverse resistance of HepG2/ADM and MCF-7/ADR cells. Anticancer Drugs 2013; 24:441-54. [DOI: 10.1097/cad.0b013e32835fcc77] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|