1
|
Phikulsod P, Sukpanichnant S, Kunacheewa C, Chieochansin T, Junking M, Yenchitsomanus PT. High prevalence of Wilms tumor 1 expression in multiple myeloma and plasmacytoma: A cohort of 142 Asian patients' samples. Pathol Oncol Res 2023; 29:1610844. [PMID: 36760714 PMCID: PMC9902379 DOI: 10.3389/pore.2023.1610844] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/26/2022] [Accepted: 01/11/2023] [Indexed: 01/25/2023]
Abstract
Wilms tumor 1 (WT1) is a promising target antigen for cancer immunotherapy. However, WT1 protein expression and its clinical correlation in multiple myeloma (MM) patients are still limited. We, therefore, investigated WT1 expression in 142 bone marrow and plasmacytoma samples of MM patients at different stages of the disease by immunohistochemistry. The correlations between WT1 expression and clinical parameters or treatment outcomes were evaluated. The overall positive rate of WT1 expression was 91.5%; this high prevalence was found in both bone marrow and plasmacytoma samples, regardless of the disease status. Cytoplasmic WT1 expression was correlated with high serum free light chain ratio at presentation. However, no significant association between WT1 expression and treatment outcome was observed. This study confirms the high prevalence of WT1 expression in an Asian cohort of MM, encouraging the development of immunotherapy targeting WT1 in MM patients, particularly in those with extramedullary plasmacytoma or relapsed disease.
Collapse
Affiliation(s)
- Ployploen Phikulsod
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand,International Graduate Program in Immunology, Department of Immunology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand,Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Sanya Sukpanichnant
- Department of Pathology, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Chutima Kunacheewa
- Division of Hematology, Department of Medicine, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Thaweesak Chieochansin
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand
| | - Mutita Junking
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand,*Correspondence: Mutita Junking, ; Pa-Thai Yenchitsomanus,
| | - Pa-Thai Yenchitsomanus
- Siriraj Center of Research Excellence for Cancer Immunotherapy (SiCORE-CIT), Division of Molecular Medicine, Research Department, Faculty of Medicine Siriraj Hospital, Mahidol University, Bangkok, Thailand,*Correspondence: Mutita Junking, ; Pa-Thai Yenchitsomanus,
| |
Collapse
|
2
|
Najar MA, Arefian M, Sidransky D, Gowda H, Prasad TSK, Modi PK, Chatterjee A. Tyrosine Phosphorylation Profiling Revealed the Signaling Network Characteristics of CAMKK2 in Gastric Adenocarcinoma. Front Genet 2022; 13:854764. [PMID: 35646067 PMCID: PMC9136244 DOI: 10.3389/fgene.2022.854764] [Citation(s) in RCA: 4] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2022] [Accepted: 03/28/2022] [Indexed: 12/24/2022] Open
Abstract
Calcium/calmodulin-dependent protein kinase kinase 2 (CAMKK2) is a serine/threonine protein kinase which functions via the calcium-triggered signaling cascade with CAMK1, CAMK4, and AMPKα as the immediate downstream substrates. CAMKK2 is reported to be overexpressed in gastric cancer; however, its signaling mechanism is poorly understood. We carried out label-free quantitative tyrosine phosphoproteomics to investigate tyrosine-mediated molecular signaling associated with CAMKK2 in gastric cancer cells. Using a high-resolution Orbitrap Fusion Tribrid Fourier-transform mass spectrometer, we identified 350 phosphotyrosine sites mapping to 157 proteins. We observed significant alterations in 81 phosphopeptides corresponding to 63 proteins upon inhibition of CAMKK2, among which 16 peptides were hyperphosphorylated corresponding to 13 proteins and 65 peptides were hypophosphorylated corresponding to 51 proteins. We report here that the inhibition of CAMKK2 leads to changes in the phosphorylation of several tyrosine kinases such as PKP2, PTK2, EPHA1, EPHA2, PRKCD, MAPK12, among others. Pathway analyses revealed that proteins are differentially phosphorylated in response to CAMKK2 inhibition involved in focal adhesions, actin cytoskeleton, axon guidance, and signaling by VEGF. The western blot analysis upon inhibition and/or silencing of CAMKK2 revealed a decrease in phosphorylation of PTK2 at Y925, c-JUN at S73, and STAT3 at Y705, which was in concordance with the mass spectrometry data. The study indicates that inhibition of CAMKK2 has an anti-oncogenic effect in gastric cells regulating phosphorylation of STAT3 through PTK2/c-JUN in gastric cancer.
Collapse
Affiliation(s)
- Mohd. Altaf Najar
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore, India
| | - Mohammad Arefian
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore, India
| | - David Sidransky
- Department of Oncology and Otolaryngology-Head and Neck Surgery, Johns Hopkins University School of Medicine, Baltimore, MD, United States
| | - Harsha Gowda
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore, India
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- Manipal Academy of Higher Education (MAHE), Manipal, India
| | - T. S. Keshava Prasad
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore, India
| | - Prashant Kumar Modi
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore, India
- *Correspondence: Prashant Kumar Modi, ; Aditi Chatterjee,
| | - Aditi Chatterjee
- Center for Systems Biology and Molecular Medicine, Yenepoya (Deemed to be University), Mangalore, India
- Institute of Bioinformatics, International Technology Park, Bangalore, India
- *Correspondence: Prashant Kumar Modi, ; Aditi Chatterjee,
| |
Collapse
|
3
|
Gao S, Zhao X, Hou L, Ma R, Zhou J, Zhu MX, Pan SJ, Li Y. The interplay between SUMOylation and phosphorylation of PKCδ facilitates oxidative stress-induced apoptosis. FEBS J 2021; 288:6447-6464. [PMID: 34089566 DOI: 10.1111/febs.16050] [Citation(s) in RCA: 3] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2021] [Revised: 04/30/2021] [Accepted: 06/03/2021] [Indexed: 12/01/2022]
Abstract
Although the increase in the number of identified posttranslational modifications (PTMs) has substantially improved our knowledge about substrate site specificity of single PTMs, the fact that different types of PTMs can crosstalk and act in concert to exert important regulatory mechanisms for protein function has not gained much attention. Here, we show that protein kinase Cδ (PKCδ) is SUMOylated at lysine 473 in its C-terminal catalytic domain, and the SUMOylation increases PKCδ stability by repressing its ubiquitination. In addition, we uncover a functional interplay between the phosphorylation and SUMOylation of PKCδ, which can strengthen each other through recruiting SUMO E2/E3 ligases and the PKCδ kinase, respectively, to the PKCδ complexes. We identified PIAS2β as the SUMO E3 ligase of PKCδ. More importantly, by enhancing PKCδ protein stability and its phosphorylation through an interdependent interplay of the PTMs, the SUMOylation of PKCδ promotes apoptotic cell death induced by H2 O2 . We conclude that SUMOylation represents an important regulatory mechanism of PKCδ PTMs for the kinase's function in oxidative cell damage.
Collapse
Affiliation(s)
- Siman Gao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, China
| | - Xiangteng Zhao
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, China
| | - Lin Hou
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, China
| | - Ruining Ma
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, China
| | - Jie Zhou
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, China
| | - Michael X Zhu
- Department of Integrative Biology and Pharmacology, McGovern Medical School, The University of Texas Health Science Center at Houston, TX, USA
| | - Si-Jian Pan
- Department of Neurosurgery, Ruijin Hospital, Shanghai Jiao Tong University School of Medicine, China
| | - Yong Li
- Department of Biochemistry and Molecular Cell Biology, Shanghai Key Laboratory for Tumor Microenvironment and Inflammation, Shanghai Jiao Tong University School of Medicine, China
| |
Collapse
|
4
|
Progress in the Development of Eukaryotic Elongation Factor 2 Kinase (eEF2K) Natural Product and Synthetic Small Molecule Inhibitors for Cancer Chemotherapy. Int J Mol Sci 2021; 22:ijms22052408. [PMID: 33673713 PMCID: PMC7957638 DOI: 10.3390/ijms22052408] [Citation(s) in RCA: 13] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2021] [Revised: 02/24/2021] [Accepted: 02/24/2021] [Indexed: 12/12/2022] Open
Abstract
Eukaryotic elongation factor 2 kinase (eEF2K or Ca2+/calmodulin-dependent protein kinase, CAMKIII) is a new member of an atypical α-kinase family different from conventional protein kinases that is now considered as a potential target for the treatment of cancer. This protein regulates the phosphorylation of eukaryotic elongation factor 2 (eEF2) to restrain activity and inhibit the elongation stage of protein synthesis. Mounting evidence shows that eEF2K regulates the cell cycle, autophagy, apoptosis, angiogenesis, invasion, and metastasis in several types of cancers. The expression of eEF2K promotes survival of cancer cells, and the level of this protein is increased in many cancer cells to adapt them to the microenvironment conditions including hypoxia, nutrient depletion, and acidosis. The physiological function of eEF2K and its role in the development and progression of cancer are here reviewed in detail. In addition, a summary of progress for in vitro eEF2K inhibitors from anti-cancer drug discovery research in recent years, along with their structure-activity relationships (SARs) and synthetic routes or natural sources, is also described. Special attention is given to those inhibitors that have been already validated in vivo, with the overall aim to provide reference context for the further development of new first-in-class anti-cancer drugs that target eEF2K.
Collapse
|
5
|
Rottlerin exhibits antitumor activity via down-regulation of TAZ in non-small cell lung cancer. Oncotarget 2018; 8:7827-7838. [PMID: 27999199 PMCID: PMC5352364 DOI: 10.18632/oncotarget.13974] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/02/2016] [Accepted: 12/09/2016] [Indexed: 02/07/2023] Open
Abstract
Rottlerin, a polyphenolic compound derived from Mallotus philipinensis, has been reported to exhibit anti-tumor activities in a variety of human malignancies including NSCLC (non-small cell lung cancer). TAZ (transcriptional co-activator with PDZ-binding motif), one of the key activators in Hippo pathway, has been characterized as an oncoprotein. Therefore, inhibition of TAZ could be useful for the treatment of human cancers. In the current study, we aimed to explore whether rottlerin inhibits the expression of TAZ in NSCLC, leading to its anti-cancer activity. Multiple approaches were applied for determining the mechanism of rottlerin-mediated anti-tumor function, including cell growth assay, Flow cytometry, wound healing assay, invasion assay, Western blotting, and transfection. We found that rottlerin inhibited cell growth, triggered apoptosis, arrested cell cycle, and retarded cell invasion in NSCLC cells. Moreover, our results showed that overexpression of TAZ enhanced cell growth, stimulated apoptosis, and promoted cell migration and invasion. Consistently, inhibition of TAZ exhibited anti-tumor activity in NSCLC cells. Notably, we validated that rottlerin exerted its tumor suppressive function via inactivation of TAZ in NSCLC cells. Taken together, our study indicates that inhibition of TAZ by rottlerin could be a promising strategy for the prevention and therapy of NSCLC.
Collapse
|
6
|
Abramson HN. Kinase inhibitors as potential agents in the treatment of multiple myeloma. Oncotarget 2018; 7:81926-81968. [PMID: 27655636 PMCID: PMC5348443 DOI: 10.18632/oncotarget.10745] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/04/2016] [Accepted: 06/30/2016] [Indexed: 12/13/2022] Open
Abstract
Recent years have witnessed a dramatic increase in the number of therapeutic options available for the treatment of multiple myeloma (MM) - from immunomodulating agents to proteasome inhibitors to histone deacetylase (HDAC) inhibitors and, most recently, monoclonal antibodies. Used in conjunction with autologous hematopoietic stem cell transplantation, these modalities have nearly doubled the disease's five-year survival rate over the last three decades to about 50%. In spite of these advances, MM still is considered incurable as resistance and relapse are common. While small molecule protein kinase inhibitors have made inroads in the therapy of a number of cancers, to date their application to MM has been less than successful. Focusing on MM, this review examines the roles played by a number of kinases in driving the malignant state and the rationale for target development in the design of a number of kinase inhibitors that have demonstrated anti-myeloma activity in both in vitro and in vivo xenograph models, as well as those that have entered clinical trials. Among the targets and their inhibitors examined are receptor and non-receptor tyrosine kinases, cell cycle control kinases, the PI3K/AKT/mTOR pathway kinases, protein kinase C, mitogen-activated protein kinase, glycogen synthase kinase, casein kinase, integrin-linked kinase, sphingosine kinase, and kinases involved in the unfolded protein response.
Collapse
Affiliation(s)
- Hanley N Abramson
- Department of Pharmaceutical Sciences, Wayne State University, Detroit, MI, USA
| |
Collapse
|
7
|
Shi J, Ning H, He G, Huang Y, Wu Z, Jin L, Jiang X. Rottlerin inhibits cell growth, induces apoptosis and cell cycle arrest, and inhibits cell invasion in human hepatocellular carcinoma. Mol Med Rep 2017; 17:459-464. [PMID: 29115596 DOI: 10.3892/mmr.2017.7924] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/06/2017] [Accepted: 08/01/2017] [Indexed: 11/06/2022] Open
Abstract
Rottlerin, a polyphenolic compound, has been demonstrated to exhibit antitumor activity in various types of human cancer. Several studies have revealed that rottlerin exerts its anticancer function through PKC‑dependent and independent pathways. The transcriptional co‑activator with PDZ‑binding motif (TAZ) oncopreotein is an important molecule in regulation of the Hippo pathway in human cancer. The present study investigated whether rottlerin has a tumor suppressive role via inhibiting the expression of TAZ, using cell viability assay, apoptosis and cell cycle analyses, western blot analysis and Tanswell invasion assay. The results demonstrated that rottlerin suppressed cell growth, triggered cell apoptosis and induced cell cycle arrest. In addition, rottlerin inhibited cell migration and invasion in hepatocellular carcinoma (HCC) cells. Mechanistically, the results demonstrated that rottlerin exerted its antitumor activity partly through the inhibition of TAZ. In addition, the depletion of TAZ led to inhibited cell growth and invasion, whereas the overexpression of TAZ enhanced cell growth and invasion in the HCC cells. Taken together, these findings indicated that the inhibition of TAZ by rottlerin may be a novel strategy for treating HCC.
Collapse
Affiliation(s)
- Jichan Shi
- Department of Infectious Disease, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Hongye Ning
- Department of Infectious Disease, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Guiqing He
- Department of Infectious Disease, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Yitong Huang
- Department of Gynecologic Oncology, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Zhengxing Wu
- Department of Infectious Disease, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Lingling Jin
- Department of Infectious Disease, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| | - Xiangao Jiang
- Department of Infectious Disease, Wenzhou Central Hospital, Wenzhou, Zhejiang 325000, P.R. China
| |
Collapse
|
8
|
Qi P, He Z, Zhang L, Fan Y, Wang Z. Rottlerin-induced autophagy leads to apoptosis in bladder cancer cells. Oncol Lett 2016; 12:4577-4583. [PMID: 28101215 PMCID: PMC5228088 DOI: 10.3892/ol.2016.5255] [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: 05/04/2015] [Accepted: 09/22/2016] [Indexed: 12/17/2022] Open
Abstract
It has been well-established that apoptosis contributes to cancer cell death; however, the role of autophagy in cancer cell death remains unclear. The aim of the present study was to investigate the effects of rottlerin, a traditional Indian medicine, on cell growth inhibition and autophagy in EJ human bladder carcinoma cells in vitro. Cell viability, measured by MTT assay, was found to be suppressed in a dose- and time-dependent manner. In addition, apoptosis was significantly increased in cells treated with rottlerin, as indicated by increased annexin V-fluorescein isothiocyanate/propidium iodide staining and changes in the cell cycle distribution that indicated blockage at G1 phase. Rottlerin treatment also enhanced the activation of autophagy, with increased expression of microtubule-associated protein 1 light chain 3 (LC3)-II and the appearance of autophagosomes. The increased level of LC3-II and autophagosomes suggests that autophagy may contribute to apoptosis in these cells. In addition, no apparent alterations in the levels of pro-caspase-3, cleaved caspase-3, total poly (ADP ribose) polymerase (PARP) and cleaved-PARP were observed in cells treated with rottlerin, which indicates that caspases may not serve a key role during the process of apoptosis induced by rottlerin. Therefore, the results of the present study indicate that rottlerin promotes apoptosis and arrests the cell cycle in EJ cells, which may be caused by autophagy activation.
Collapse
Affiliation(s)
- Ping Qi
- Institute of Urology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China; Department of Clinical Laboratory, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Zhenhua He
- Institute of Urology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China; Neurosurgery, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Lixiu Zhang
- Institute of Urology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China
| | - Yuan Fan
- The Second School of Clinical Medicine, Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| | - Zhiping Wang
- Institute of Urology, Lanzhou University Second Hospital, Lanzhou, Gansu 730030, P.R. China; Key Laboratory of Urological Diseases in Gansu Province, Lanzhou University, Lanzhou, Gansu 730030, P.R. China
| |
Collapse
|
9
|
Ashour AA, Abdel-Aziz AAH, Mansour AM, Alpay SN, Huo L, Ozpolat B. Targeting elongation factor-2 kinase (eEF-2K) induces apoptosis in human pancreatic cancer cells. Apoptosis 2014; 19:241-58. [PMID: 24193916 DOI: 10.1007/s10495-013-0927-2] [Citation(s) in RCA: 63] [Impact Index Per Article: 6.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Pancreatic cancer (PaCa) is one of the most aggressive, apoptosis-resistant and currently incurable cancers with a poor survival rate. Eukaryotic elongation factor-2 kinase (eEF-2K) is an atypical kinase, whose role in PaCa survival is not yet known. Here, we show that eEF-2K is overexpressed in PaCa cells and its down-regulation induces apoptotic cell death. Rottlerin (ROT), a polyphenolic compound initially identified as a PKC-δ inhibitor, induces apoptosis and autophagy in a variety of cancer cells including PaCa cells. We demonstrated that ROT induces intrinsic apoptosis, with dissipation of mitochondrial membrane potential (ΔΨm), and stimulates extrinsic apoptosis with concomitant induction of TNF-related apoptosis inducing ligand (TRAIL) receptors, DR4 and DR5, with caspase-8 activation, in PANC-1 and MIAPaCa-2 cells. Notably, while none of these effects were dependent on PKC-δ inhibition, ROT down-regulates eEF-2K at mRNA level, and induce eEF-2K protein degradation through ubiquitin-proteasome pathway. Down-regulation of eEF-2K recapitulates the events observed after ROT treatment, while its over-expression suppressed the ROT-induced apoptosis. Furthermore, eEF-2K regulates the expression of tissue transglutaminase (TG2), an enzyme previously implicated in proliferation, drug resistance and survival of cancer cells. Inhibition of eEF-2K/TG2 axis leads to caspase-independent apoptosis which is associated with induction of apoptosis-inducing factor (AIF). Collectively, these results indicate, for the first time, that the down-regulation of eEF-2K leads to induction of intrinsic, extrinsic as well as AIF-dependent apoptosis in PaCa cells, suggesting that eEF-2K may represent an attractive therapeutic target for the future anticancer agents in PaCa.
Collapse
Affiliation(s)
- Ahmed A Ashour
- Department of Experimental Therapeutics, Unit 422, The University of Texas, M.D. Anderson Cancer Center, 1515 Holcombe Boulevard, Houston, TX, 77030, USA
| | | | | | | | | | | |
Collapse
|
10
|
Ikewaki N, Kulski JK, Inoko H. Regulation of CD93 Cell Surface Expression by Protein Kinase C Isoenzymes. Microbiol Immunol 2013; 50:93-103. [PMID: 16490927 DOI: 10.1111/j.1348-0421.2006.tb03774.x] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/26/2022]
Abstract
Human CD93, also known as complement protein 1, q subcomponent, receptor (C1qRp), is selectively expressed by cells with a myeloid lineage, endothelial cells, platelets, and microglia and was originally reported to be involved in the complement protein 1, q subcomponent (C1q)-mediated enhancement of phagocytosis. The intracellular molecular events responsible for the regulation of its expression on the cell surface, however, have not been determined. In this study, the effect of protein kinases in the regulation of CD93 expression on the cell surface of a human monocyte-like cell line (U937), a human NK-like cell line (KHYG-1), and a human umbilical vein endothelial cell line (HUV-EC-C) was investigated using four types of protein kinase inhibitors, the classical protein kinase C (cPKC) inhibitor Go6976, the novel PKC (nPKC) inhibitor Rottlerin, the protein kinase A (PKA) inhibitor H-89 and the protein tyrosine kinase (PTK) inhibitor herbimycin A at their optimum concentrations for 24 hr. CD93 expression was analyzed using flow cytometry and glutaraldehyde-fixed cellular enzyme-linked immunoassay (EIA) techniques utilizing a CD93 monoclonal antibody (mAb), mNI-11, that was originally established in our laboratory as a CD93 detection probe. The nPKC inhibitor Rottlerin strongly down-regulated CD93 expression on the U937 cells in a dose-dependent manner, whereas the other inhibitors had little or no effect. CD93 expression was down-regulated by Go6976, but not by Rottlerin, in the KHYG-1 cells and by both Rottlerin and Go6976 in the HUV-EC-C cells. The PKC stimulator, phorbol myristate acetate (PMA), strongly up-regulated CD93 expression on the cell surface of all three cell-lines and induced interleukin-8 (IL-8) production by the U937 cells and interferon-gamma (IFN-gamma) production by the KHYG-1 cells. In addition, both Go6976 and Rottlerin inhibited the up-regulation of CD93 expression induced by PMA and IL-8 or IFN-gamma production in the respective cell-lines. Whereas recombinant tumor necrosis factor-alpha (rTNF-alpha) slightly up-regulated CD93 expression on the U937 cells, recombinant interleukin-1beta (rIL-1beta), recombinant interleukin-2 (rIL-2), recombinant interferon-gamma (rIFN-gamma) and lipopolysaccharide (LPS) had no effect. Taken together, these findings indicate that the regulation of CD93 expression on these cells involves the PKC isoenzymes.
Collapse
Affiliation(s)
- Nobunao Ikewaki
- Institute of Immunology, Kyushu University of Health and Welfare, Nobeoka, Miyazaki, Japan
| | | | | |
Collapse
|
11
|
Kiaii S, Kokhaei P, Mozaffari F, Rossmann E, Pak F, Moshfegh A, Palma M, Hansson L, Mashayekhi K, Hojjat-Farsangi M, Österborg A, Choudhury A, Mellstedt H. T cells from indolent CLL patients prevent apoptosis of leukemic B cells in vitro and have altered gene expression profile. Cancer Immunol Immunother 2013; 62:51-63. [PMID: 22736254 PMCID: PMC11029037 DOI: 10.1007/s00262-012-1300-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2011] [Accepted: 05/30/2012] [Indexed: 10/28/2022]
Abstract
T cells may have a role in sustaining the leukemic clone in chronic lymphocytic leukemia (CLL). In this study, we have examined the ability of T cells from CLL patients to support the survival of the leukemic B cells in vitro. Additionally, we compared global gene expression of T cells from indolent CLL patients with healthy individuals and multiple myeloma (MM) patients. Apoptosis of purified leukemic B cells was inhibited in vitro when co-cultured with increasing numbers of autologous T cells (p < 0.01) but not autologous B and T cells of normal donors. The anti-apoptotic effect exceeded that of the anti-apoptotic cytokine IL-4 (p = 0.002) and was greater with CD8+ cells (p = 0.02) than with CD4+ cells (p = 0.05). The effect was depended mainly on cell-cell contact although a significant effect was also observed in transwell experiments (p = 0.05). About 356 genes involved in different cellular pathways were deregulated in T cells of CLL patients compared to healthy individuals and MM patients. The results of gene expression profiling were verified for 6 genes (CCL4, CCL5 (RANTES), XCL1, XCL2, KLF6, and TRAF1) using qRT-PCR and immunoblotting. Our results demonstrate that CLL-derived T cells can prevent apoptosis of leukemic B cells and have altered expression of genes that may facilitate the survival of the leukemic clone.
Collapse
MESH Headings
- Aged
- Aged, 80 and over
- Apoptosis/immunology
- B-Lymphocytes/immunology
- Coculture Techniques
- Female
- Flow Cytometry
- Humans
- Immunoblotting
- Leukemia, Lymphocytic, Chronic, B-Cell/genetics
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Male
- Middle Aged
- Real-Time Polymerase Chain Reaction
- Reverse Transcriptase Polymerase Chain Reaction
- T-Lymphocytes/immunology
- Transcriptome
Collapse
Affiliation(s)
- Shahryar Kiaii
- Institute of Cancer, Barts and The London School of Medicine and Dentistry, Charterhouse Square, London, EC1M 6BQ UK
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Parviz Kokhaei
- Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Fariba Mozaffari
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Eva Rossmann
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska University Hospital, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Fatemeh Pak
- Department of Immunology, Semnan University of Medical Sciences, Semnan, Iran
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Ali Moshfegh
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Marzia Palma
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska University Hospital, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Lotta Hansson
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska University Hospital, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Kaveh Mashayekhi
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Mohammad Hojjat-Farsangi
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Anders Österborg
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska University Hospital, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
- Department of Hematology, Karolinska University Hospital, Stockholm, Sweden
| | - Aniruddha Choudhury
- Center for Immune and Targeted Therapy Greenslopes Private Hospital, Newdegate Street, Brisbane, QLD 4072 Australia
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska Institutet, 171 76 Stockholm, Sweden
| | - Håkan Mellstedt
- Cancer Center Karolinska, Department of Oncology-Pathology (Radiumhemmet), Karolinska University Hospital, Solna, Karolinska Institutet, 171 76 Stockholm, Sweden
| |
Collapse
|
12
|
Zhao S, Ma CM, Liu CX, Wei W, Sun Y, Yan H, Wu YL. Autophagy inhibition enhances isobavachalcone-induced cell death in multiple myeloma cells. Int J Mol Med 2012; 30:939-44. [PMID: 22824846 DOI: 10.3892/ijmm.2012.1066] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2012] [Accepted: 06/13/2012] [Indexed: 11/06/2022] Open
Abstract
Despite recent advancements in therapeutic drugs, multiple myeloma remains an incurable disease. Therefore, a more effective treatment is urgently required. In this study, we show that isobavachalcone (IBC), a natural chalcone compound, induces apoptosis- and autophagy-related cell death in myeloma cells. The inhibition of autophagy by knocking down beclin-1 or by using autophagy inhibitors, such as 3-methyladenine, bafilomycin A and chloroquine significantly enhanced IBC-induced cell death, as demonstrated by the increased number of Annexin V-positive cells. Moreover, we demonstrate that the collapse of the mitochondrial membrane potential contributes to chloroquine and IBC-induced cell death, which is accompanied by the activation of caspase-9, and -3, the cleavage of poly (ADP-ribose) polymerase (PARP) and the proteolytic activation of protein kinase Cδ (PKCδ). Furthermore, the inhibition of the activation of PKCδ by rottlerin, an inhibitor of PKCδ, not only suppressed the activation of PKCδ, but also the apoptosis induced by the co-treatment of chloroquine and IBC, indicating the involvement of PKCδ in chloroquine plus IBC-induced cell death. Finally, the combination of chloroquine and IBC had little effect on the viability of normal peripheral blood mononuclear cells. As both chloroquine and IBC have been shown to be relatively specific for cancer cells, the combination of these two agents at non-toxic or sub-toxic concentrations represents an attractive novel regimen for myeloma treatment and warrants further investigation in preclinical and clinical studies.
Collapse
Affiliation(s)
- Shan Zhao
- Department of Pathophysiology, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of National Ministry of Education, Shanghai 200025, PR China
| | | | | | | | | | | | | |
Collapse
|
13
|
Zhang J, Chen F, Li W, Xiong Q, Yang M, Zheng P, Li C, Pei J, Ge F. 14-3-3ζ interacts with stat3 and regulates its constitutive activation in multiple myeloma cells. PLoS One 2012; 7:e29554. [PMID: 22279540 PMCID: PMC3261159 DOI: 10.1371/journal.pone.0029554] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2011] [Accepted: 11/30/2011] [Indexed: 12/18/2022] Open
Abstract
The 14-3-3 proteins are a family of regulatory signaling molecules that interact with other proteins in a phosphorylation-dependent manner and function as adapter or scaffold proteins in signal transduction pathways. One family member, 14-3-3ζ, is believed to function in cell signaling, cycle control, and apoptotic death. A systematic proteomic analysis done in our laboratory has identified signal transducers and activators of transcription 3 (Stat3) as a novel 14-3-3ζ interacting protein. Following our initial finding, in this study, we provide evidence that 14-3-3ζ interacts physically with Stat3. We further demonstrate that phosphorylation of Stat3 at Ser727 is vital for 14-3-3ζ interaction and mutation of Ser727 to Alanine abolished 14-3-3ζ/Stat3 association. Inhibition of 14-3-3ζ protein expression in U266 cells inhibited Stat3 Ser727 phosphorylation and nuclear translocation, and decreased both Stat3 DNA binding and transcriptional activity. Moreover, 14-3-3ζ is involved in the regulation of protein kinase C (PKC) activity and 14-3-3ζ binding to Stat3 protects Ser727 dephosphorylation from protein phosphatase 2A (PP2A). Taken together, our findings support the model that multiple signaling events impinge on Stat3 and that 14-3-3ζ serves as an essential coordinator for different pathways to regulate Stat3 activation and function in MM cells.
Collapse
Affiliation(s)
- Jia Zhang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Fangjin Chen
- Center for Theoretical Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
| | - Wenliang Li
- School of Science and Technology, Tokai University, Tokyo, Japan
| | - Qian Xiong
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Mingkun Yang
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Peng Zheng
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Chongyang Li
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
| | - Jianfeng Pei
- Center for Theoretical Biology, Academy for Advanced Interdisciplinary Studies, Peking University, Beijing, China
- * E-mail: (JP); (FG)
| | - Feng Ge
- Institute of Hydrobiology, Chinese Academy of Sciences, Wuhan, China
- * E-mail: (JP); (FG)
| |
Collapse
|
14
|
Maioli E, Torricelli C, Valacchi G. Rottlerin and cancer: novel evidence and mechanisms. ScientificWorldJournal 2012; 2012:350826. [PMID: 22272173 PMCID: PMC3259573 DOI: 10.1100/2012/350826] [Citation(s) in RCA: 47] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2011] [Accepted: 11/14/2011] [Indexed: 12/26/2022] Open
Abstract
Because cancers are caused by deregulation of hundreds of genes, an ideal anticancer agent should target multiple gene products or signaling pathways simultaneously. Recently, extensive research has addressed the chemotherapeutic potential of plant-derived compounds. Among the ever-increasing list of naturally occurring anticancer agents, Rottlerin appears to have great potentiality for being used in chemotherapy because it affects several cell machineries involved in survival, apoptosis, autophagy, and invasion. The underlying mechanisms that have been described are diverse, and the final, cell-specific, Rottlerin outcome appears to result from a combination of signaling pathways at multiple levels. This paper seeks to summarize the multifocal signal modulatory properties of Rottlerin, which merit to be further exploited for successful prevention and treatment of cancer.
Collapse
Affiliation(s)
- E Maioli
- Department of Physiology, University of Siena, Aldo Moro Street, 53100 Siena, Italy.
| | | | | |
Collapse
|
15
|
Zhu L, Song S, Pi Y, Yu Y, She W, Ye H, Su Y, Hu Q. Cumulated Ca2+ spike duration underlies Ca2+ oscillation frequency-regulated NFκB transcriptional activity. J Cell Sci 2011; 124:2591-601. [DOI: 10.1242/jcs.082727] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/25/2023] Open
Abstract
[Ca2+]i oscillations drive downstream events, like transcription, in a frequency-dependent manner. Why [Ca2+]i oscillation frequency regulates transcription has not been clearly revealed. A variation in [Ca2+]i oscillation frequency apparently leads to a variation in the time duration of cumulated [Ca2+]i elevations or cumulated [Ca2+]i spike duration. By manipulating [Ca2+]i spike duration, we generated a series of [Ca2+]i oscillations with the same frequency but different cumulated [Ca2+]i spike durations, as well as [Ca2+]i oscillations with the different frequencies but the same cumulated [Ca2+]i spike duration. Molecular assays demonstrated that, when generated in ‘artificial’ models alone, under physiologically simulated conditions or repetitive pulses of agonist exposure, [Ca2+]i oscillation regulates NFκB transcriptional activity, phosphorylation of IκBα and Ca2+-dependent gene expression all in a way actually dependent on cumulated [Ca2+]i spike duration whether or not frequency varies. This study underlines that [Ca2+]i oscillation frequency regulates NFκB transcriptional activity through cumulated [Ca2+]i spike-duration-mediated IκBα phosphorylation.
Collapse
Affiliation(s)
- Liping Zhu
- Department of Pathophysiology, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, People's Republic of China
- Key Laboratory of Pulmonary Diseases of Ministry of Health of China, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, People's Republic of China
| | - Shanshan Song
- Department of Pathophysiology, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, People's Republic of China
- Key Laboratory of Pulmonary Diseases of Ministry of Health of China, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, People's Republic of China
| | - Yubo Pi
- Department of Pathophysiology, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, People's Republic of China
- Key Laboratory of Pulmonary Diseases of Ministry of Health of China, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, People's Republic of China
| | - Yang Yu
- Department of Pathophysiology, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, People's Republic of China
- Key Laboratory of Pulmonary Diseases of Ministry of Health of China, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, People's Republic of China
| | - Weibin She
- Department of Pathophysiology, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, People's Republic of China
- Key Laboratory of Pulmonary Diseases of Ministry of Health of China, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, People's Republic of China
| | - Hong Ye
- Department of Pathophysiology, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, People's Republic of China
- Key Laboratory of Pulmonary Diseases of Ministry of Health of China, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, People's Republic of China
| | - Yuan Su
- Key Laboratory of Pulmonary Diseases of Ministry of Health of China, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, People's Republic of China
- Department of Respiratory Medicine, Union Hospital, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, People's Republic of China
| | - Qinghua Hu
- Department of Pathophysiology, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, People's Republic of China
- Key Laboratory of Pulmonary Diseases of Ministry of Health of China, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, People's Republic of China
- The MOE Key Laboratory of Environment and Health, School of Public Health, Tongji Medical College, Huazhong Science and Technology University, Wuhan 430030, People's Republic of China
| |
Collapse
|
16
|
Ohno I, Eibl G, Odinokova I, Edderkaoui M, Damoiseaux RD, Yazbec M, Abrol R, Goddard WA, Yokosuka O, Pandol SJ, Gukovskaya AS. Rottlerin stimulates apoptosis in pancreatic cancer cells through interactions with proteins of the Bcl-2 family. Am J Physiol Gastrointest Liver Physiol 2010; 298:G63-73. [PMID: 19762431 PMCID: PMC2806098 DOI: 10.1152/ajpgi.00257.2009] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
Rottlerin is a polyphenolic compound derived from Mallotus philipinensis. In the present study, we show that rottlerin decreased tumor size and stimulated apoptosis in an orthotopic model of pancreatic cancer with no effect on normal tissues in vivo. Rottlerin also induced apoptosis in pancreatic cancer (PaCa) cell lines by interacting with mitochondria and stimulating cytochrome c release. Immunoprecipitation results indicated that rottlerin disrupts complexes of prosurvival Bcl-xL with Bim and Puma. Furthermore, siRNA knockdown showed that Bim and Puma are necessary for rottlerin to stimulate apoptosis. We also showed that rottlerin and Bcl-2 and Bcl-xL inhibitor BH3I-2' stimulate apoptosis through a common mechanism. They both directly interact with mitochondria, causing increased cytochrome c release and mitochondrial depolarization, and both decrease sequestration of BH3-only proteins by Bcl-xL. However, the effects of rottlerin and BH3I-2' on the complex formation between Bcl-xL and BH3-only proteins are different. BH3I-2' disrupts complexes of Bcl-xL with Bad but not with Bim or Puma, whereas rottlerin had no effect on the Bcl-xL interaction with Bad. Also BH3I-2', but not rottlerin, required Bad to stimulate apoptosis. In conclusion, our results demonstrate that rottlerin has a potent proapoptotic and antitumor activity in pancreatic cancer, which is mediated by disrupting the interaction between prosurvival Bcl-2 proteins and proapoptotic BH3-only proteins. Thus rottlerin represents a promising novel agent for pancreatic cancer treatment.
Collapse
Affiliation(s)
- Izumi Ohno
- 1Veterans Affairs Greater Los Angeles Healthcare System, ,Departments of 2Medicine and ,7Department of Medicine and Clinical Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Guido Eibl
- 3Surgery, David Geffen School of Medicine, and
| | - Irina Odinokova
- 1Veterans Affairs Greater Los Angeles Healthcare System, ,Departments of 2Medicine and ,4Institute of Theoretical and Experimental Biophysics, Pushchino, Russia;
| | - Mouad Edderkaoui
- 1Veterans Affairs Greater Los Angeles Healthcare System, ,Departments of 2Medicine and
| | - Robert D. Damoiseaux
- 5Molecular Shared Screening Resources, University of California, Los Angeles, California;
| | - Moussa Yazbec
- 1Veterans Affairs Greater Los Angeles Healthcare System, ,Departments of 2Medicine and
| | - Ravinder Abrol
- 6Materials and Process Simulation Center, Beckman Institute, California Institute of Technology, Pasadena, California;
| | - William A. Goddard
- 6Materials and Process Simulation Center, Beckman Institute, California Institute of Technology, Pasadena, California;
| | - Osamu Yokosuka
- 7Department of Medicine and Clinical Oncology, Graduate School of Medicine, Chiba University, Chiba, Japan
| | - Stephen J. Pandol
- 1Veterans Affairs Greater Los Angeles Healthcare System, ,Departments of 2Medicine and
| | - Anna S. Gukovskaya
- 1Veterans Affairs Greater Los Angeles Healthcare System, ,Departments of 2Medicine and
| |
Collapse
|
17
|
Abstract
Emerging evidence indicates that suppression of protein kinase C (PKC) renders the susceptibility of cells expressing mutated ras to apoptosis. Although the effort has been made, the underlying molecular mechanisms are not fully understood. In this study, using small hairpin RNAs (shRNAs) or PKC inhibitor, we show that the concurrent suppression of PKC-alpha and beta induces cells ectopically expressing v-ras to undergo apoptosis. In this apoptotic process, PKC-delta is upregulated and translocated from the cytosol to the nucleus. The activated PKC-delta associates with and phosphorylates p73 to initiate apoptosis. In this apoptotic process, Akt seems to be downstream of oncogenic Ras. Moreover, overexpression of PKC-delta, without co-suppression of PKC-alpha and beta, is not apoptotic to the cells, suggesting that PKC-delta and PKC-alpha/beta function oppositely to facilitate cells harboring v-ras to survive. Thus, our study shows that PKC-alpha and beta are necessary for sustaining the homeostasis in cells containing a hyperactive Ras. The abrogation of these two isoforms switches on the p73-regulated apoptotic machinery through the activation of PKC-delta.
Collapse
|
18
|
Lu ZG, Liu H, Yamaguchi T, Miki Y, Yoshida K. Protein kinase Cdelta activates RelA/p65 and nuclear factor-kappaB signaling in response to tumor necrosis factor-alpha. Cancer Res 2009; 69:5927-35. [PMID: 19549902 DOI: 10.1158/0008-5472.can-08-4786] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
Nuclear factor-kappaB (NF-kappaB) is tightly modulated by IkappaB kinases and IkappaBalpha in the cytoplasm. On stimulation, NF-kappaB translocates into the nucleus to initiate transcription; however, regulation of its transcriptional activity remains obscure. Here, we show that protein kinase C (PKC) delta controls the main subunit of NF-kappaB, RelA/p65. On exposure to tumor necrosis factor-alpha (TNF-alpha), the expression of RelA/p65 target genes such as IkappaBalpha, RelB, and p100/p52 is up-regulated in a PKCdelta-dependent manner. The results also show that PKCdelta is targeted to the nucleus and forms a complex with RelA/p65 following TNF-alpha exposure. Importantly, kinase activity of PKCdelta is required for RelA/p65 transactivation. In concert with these results, PKCdelta activates RelA/p65 for its occupancy to target-gene promoters, including IkappaBalpha and p100/p52. Moreover, functional analyses show that inhibition of PKCdelta is associated with substantial attenuation of NF-kappaB activity in response to TNF-alpha. These findings provide evidence that PKCdelta orchestrates RelA/p65 transactivation, a requisite for NF-kappaB signaling pathway in the nucleus.
Collapse
Affiliation(s)
- Zheng-Guang Lu
- Department of Molecular Genetics, Medical Research Institute, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | |
Collapse
|
19
|
Integrin signaling and cell spreading alterations by rottlerin treatment of chick limb bud mesenchymal cells. Biochimie 2009; 91:624-31. [DOI: 10.1016/j.biochi.2009.03.006] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2009] [Accepted: 03/12/2009] [Indexed: 11/21/2022]
|
20
|
Ikewaki N, Fujii N, Onaka T, Ikewaki S, Inoko H. Immunological actions of Sophy beta-glucan (beta-1,3-1,6 glucan), currently available commercially as a health food supplement. Microbiol Immunol 2008; 51:861-73. [PMID: 17895603 DOI: 10.1111/j.1348-0421.2007.tb03982.x] [Citation(s) in RCA: 58] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
We examined the immunological actions of Sophy beta-glucan(Ikewaki N., et al. United States Patent 6956120 and Japan Patent 2004-329077), a type of beta-1,3-1,6 glucan produced by the black yeast Aureobasidium pullulans (A. pullulans) strain AFO-202, currently available commercially as a health food supplement, using different human in vitro experimental systems. Sophy beta-glucan significantly (P<0.01) stimulated the (3)H-thymidine incorporation rates (marker of DNA synthesis) in human peripheral blood mononuclear cells (PBMCs) obtained from normal adult donors, in vitro. Enzyme-linked immunoassays (EIAs) revealed that Sophy beta-glucan stimulated the production of interleukin-8 (IL-8) or soluble Fas (sFas), but not that of IL-1beta, IL-2, IL-6, IL-12 (p70+40), interferon-gamma (IFN-gamma), tumor necrosis factor-alpha (TNF-alpha) or soluble Fas ligand (sFasL), in either cultured PBMCs or cells of the human monocyte-like cell line, U937. The induction by Sophy beta-glucan of DNA synthesis in PBMCs was completely blocked by the addition of monoclonal antibodies (mAbs) to CD11a, CD54, human leukocyte antigen-class II (HLA-class II), Toll-like receptor-2 (TLR-2), and Toll-like receptor-4 (TLR-4). In these blocking experiments using the mAbs, the main differences in the results between PBMCs and U937 cells were that the mAbs against TLR-2 and TLR-4 did not block the Sophy beta-glucan-induced production of IL-8 in the U937 cells. Furthermore, a mAb to the beta-glucan receptor, Dectin-1, significantly (P<0.05) blocked the Sophy beta-glucan induced DNA synthesis in the PBMCs, and Sophy beta-glucan-induced production of IL-8 in the U937 cells. The Sophy beta-glucan-induced production of IL-8 in the U937 cells was significantly (P<0.01) blocked by the conventional protein kinase C (PKC) inhibitor Go6976, the novel PKC inhibitor Rottlerin, the protein kinase A (PKA) inhibitor H-89, and the protein tyrosine kinase (PTK) inhibitor herbimycin A. Among these, the blocking effect of the novel PKC (PKC delta isoenzyme) inhibitor Rottlerin was the most pronounced. Studies employing reverse transcriptase-polymerase chain reaction (RT-PCR) showed that Sophy beta-glucan stimulated the expression of IL-8 mRNA in the U937 cells, and that this induction was inhibited by Rottlerin. Sophy beta-glucan also blocked the stimulator cell induction of DNA synthesis and IFN-gamma production in the responder cells in a one-way mixed lymphocyte reaction (MLR) using allogenic PBMCs. Interestingly, immunoglobulin G (IgG), but not IgM to Sophy beta-glucan was detected in the sera derived from normal adult donors and from the umbilical cord blood of neonates. Taken together, these findings strongly suggest that the Sophy beta-glucan may have unique immune regulatory or enhancing properties that could be exploited by the health food, medical and pharmaceutical industries.
Collapse
Affiliation(s)
- Nobunao Ikewaki
- Kyushu University of Health and Welfare School of Health Science, and Institute of Immunology, Takahashi Educational Institute, Nobeoka, Miyazaki, Japan.
| | | | | | | | | |
Collapse
|
21
|
Baumann P, Armann J, Mandl-Weber S, Grün G, Oduncu F, Schmidmaier R. Inhibitors of protein kinase C sensitise multiple myeloma cells to common genotoxic drugs. Eur J Haematol 2007; 80:37-45. [PMID: 18028419 DOI: 10.1111/j.1600-0609.2007.00977.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
OBJECTIVES Despite high dose treatment regimes multiple myeloma (MM) disease is still not curable. Patients become resistant to cytotoxic drugs and die of disease progression. Therefore, besides new cytotoxic compounds drug sensitisers are urgently needed. METHODS The MM cell lines U266, OPM-2, RPMI-8226 and NCI-H929 were incubated with the common anti-myeloma drugs like melphalan together with protein kinase C (PKC) inhibitors. Growth inhibition was measured using the water-soluble tetrazolium salt 4-[3-(4-iodophenyl)-2-(4-nitrophenyl)-2H-5-tetrazolio]-1,3-benzene disulfonate (WST-1 assay), and apoptosis was determined by flow cytometry after staining with fluorescein isothiocyanate-labeled annexin V (annexin-V-FITC) and propidium iodide. Intracellular signalling was shown by western blotting. RESULTS In this study we show that the combination of melphalan or doxorubicin with a PKC inhibitor, Gö6976 or enzastaurin, strongly increases cell toxicity. Increase of cytotoxicity is shown to be due to increased induction of apoptosis. Furthermore, we show that the protective effect of human bone marrow stromal cells (hBMSC) is abrogated by the PKC inhibitors. Finally, western blotting experiments revealed that incubation of myeloma cells with cytotoxic drugs like melphalan or doxorubicin leads to increased phosphorylation and therefore degradation of inhibitor of nuclear factor kappa B (IkappaB) and release of nuclear factor kappa B (NFkappaB). In contrast, enzastaurin inhibits phosphorylation of IkappaB. CONCLUSIONS We conclude that the combination of conventional drugs and PKC inhibitors might be very effective and represents a new strategy in the treatment of MM.
Collapse
Affiliation(s)
- Philipp Baumann
- Department of Hematology and Oncology, Medizinische Klinik Innenstadt, Klinikum der Universität München, Germany.
| | | | | | | | | | | |
Collapse
|
22
|
Podar K, Raab MS, Chauhan D, Anderson KC. The therapeutic role of targeting protein kinase C in solid and hematologic malignancies. Expert Opin Investig Drugs 2007; 16:1693-707. [PMID: 17922632 DOI: 10.1517/13543784.16.10.1693] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The protein kinase C (PKC) family, the most prominent target of tumor-promoting phorbol esters, is functionally linked to cell differentiation, growth, survival, migration and tumorigenesis and so mediates tumor cell proliferation, survival, multidrug resistance, invasion, metastasis and tumor angiogenesis. Therefore, targeting PKC isozymes may represent an attractive target for novel anticancer therapies. Recent preclinical and clinical studies using the macrocyclic bisindolylmaleimide enzastaurin or the N-benzylstaurosporine midostaurin demonstrate promising activity of PKC inhibitors in a variety of tumors, including diffuse large B-cell lymphoma, multiple myeloma and Waldenstroem's macroglobulinemia. However, our knowledge of PKCs in tumorigenesis is still only partial and each PKC isoform may contribute to tumorigenesis in a distinct way. Specifically, PKC isoforms have vastly different roles, which vary depending on expression levels of organ and tissue distribution, cell type, intracellular localization, protein-protein and lipid-protein interactions and the biologic environment. Although PKC activation generally positively affects tumor cell growth, motility, invasion and metastasis, recent reports show that many PKCs can also have negative effects. Therefore, it is necessary to further dissect the relative contribution of PKC isozymes in the development and progression of specific tumors in order to identify therapeutic opportunities, using either PKC inhibitors or PKC activators.
Collapse
Affiliation(s)
- Klaus Podar
- Dana-Farber Cancer Institute, Department of Medical Oncology, Jerome Lipper Multiple Myeloma Center, 44 Binney Street, Boston, MA 02115, USA.
| | | | | | | |
Collapse
|
23
|
|
24
|
Kurosu T, Tsuji K, Kida A, Koyama T, Yamamoto M, Miura O. Rottlerin synergistically enhances imatinib-induced apoptosis of BCR/ABL-expressing cells through its mitochondrial uncoupling effect independent of protein kinase C-δ. Oncogene 2006; 26:2975-87. [PMID: 17130834 DOI: 10.1038/sj.onc.1210117] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Although the BCR/ABL tyrosine kinase inhibitor imatinib is highly effective for treatment of chronic myeloid leukemia (CML) and Philadelphia-chromosome positive acute lymphoblastic leukemia (ALL), relapse with emerging imatinib-resistance mutations in the BCR/ABL kinase domain poses a significant problem. Here, we demonstrate that rottlerin, a putative protein kinase C-delta (PKCdelta)-specific inhibitor, acts synergistically with imatinib to induce apoptosis of BCR/ABL-expressing K562 and Ton.B210 cells. However, rottlerin inhibited neither PKCdelta nor BCR/ABL in these cells. On the other hand, rottlerin, previously characterized also as a mitochondrial uncoupler, transiently but significantly reduced mitochondrial membrane potential and gradually induced mitochondrial membrane permeabilization. Moreover, two other mitochondrial uncouplers, FCCP and DNP, very similarly induced apoptosis of BCR/ABL-expressing cells in a synergistic manner with imatinib. Imatinib synergistically enhanced mitochondrial membrane permeabilization induced by mitochondrial uncouplers, which led to release of cytochrome c into the cytoplasm and activation of caspases-3 and -9. Rottlerin also enhanced the cytotoxic effect of imatinib in leukemic cells from patients with CML blast crisis and Ph-positive ALL or a cell line expressing the imatinib-resistant E255K BCR/ABL mutant. The present study indicates that rottlerin synergistically enhances imatinib-induced apoptosis through its mitochondrial uncoupling effect independent of PKCdelta and may contribute to the development of new treatment strategy to overcome the imatinib resistance and to cure the BCR/ABL expressing leukemias.
Collapse
Affiliation(s)
- T Kurosu
- Department of Hematology, Graduate School of Medicine, Tokyo Medical and Dental University, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
25
|
Podar K, Raab MS, Zhang J, McMillin D, Breitkreutz I, Tai YT, Lin BK, Munshi N, Hideshima T, Chauhan D, Anderson KC. Targeting PKC in multiple myeloma: in vitro and in vivo effects of the novel, orally available small-molecule inhibitor enzastaurin (LY317615.HCl). Blood 2006; 109:1669-77. [PMID: 17023575 PMCID: PMC1794057 DOI: 10.1182/blood-2006-08-042747] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/24/2023] Open
Abstract
In multiple myeloma (MM) protein kinase C (PKC) signaling pathways have been implicated in cell proliferation, survival, and migration. Here we investigated the novel, orally available PKC-inhibitor enzastaurin for its anti-MM activity. Enzastaurin specifically inhibits phorbol ester-induced activation of PKC isoforms, as well as phosphorylation of downstream signaling molecules MARCKS and PKCmu. Importantly, it also inhibits PKC activation triggered by growth factors and cytokines secreted by bone marrow stromal cells (BMSCs), costimulation with fibronectin, vascular endothelial growth factor (VEGF), or interleukin-6 (IL-6), as well as MM patient serum. Consequently, enzastaurin inhibits proliferation, survival, and migration of MM cell lines and MM cells isolated from multidrug-resistant patients and overcomes MM-cell growth triggered by binding to BMSCs and endothelial cells. Importantly, strong synergistic cytotoxicity is observed when enzastaurin is combined with bortezomib and moderate synergistic or additive effects when combined with melphalan or lenalidomide. Finally, tumor growth, survival, and angiogenesis are abrogated by enzastaurin in an in vivo xenograft model of human MM. Our results therefore demonstrate in vitro and in vivo efficacy of the orally available PKC inhibitor enzastaurin in MM and strongly support its clinical evaluation, alone or in combination therapies, to improve outcome in patients with MM.
Collapse
Affiliation(s)
- Klaus Podar
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Rizvi MA, Ghias K, Davies KM, Ma C, Weinberg F, Munshi HG, Krett NL, Rosen ST. Enzastaurin (LY317615), a protein kinase Cβ inhibitor, inhibits the AKT pathway and induces apoptosis in multiple myeloma cell lines. Mol Cancer Ther 2006; 5:1783-9. [PMID: 16891464 DOI: 10.1158/1535-7163.mct-05-0465] [Citation(s) in RCA: 64] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Enzastaurin (LY317615), an acyclic bisindolylmaleimide, is an oral inhibitor of the protein kinase Cbeta isozyme. The objective of this study was to assess the efficacy of enzastaurin in inducing apoptosis in multiple myeloma (MM) cell lines and to investigate possible mechanisms of apoptosis. Cell proliferation assays were done on a variety of MM cell lines with unique characteristics (dexamethasone sensitive, dexamethasone resistant, chemotherapy sensitive, and melphalan resistant). The dexamethasone-sensitive MM.1S cell line was used to further assess the effect of enzastaurin in the presence of dexamethasone, insulin-like growth factor-I (IGF-I), interleukin-6, and the pan-specific caspase inhibitor ZVAD-fmk. Enzastaurin increased cell death in all cell lines at clinically significant low micromolar concentrations (1-3 micromol/L) after 72 hours of treatment. Dexamethasone and enzastaurin were shown to have an additive effect on MM.1S cell death. Although IGF-I blocked the effect of 1 micromol/L enzastaurin, IGF-I did not abrogate cell death induced with 3 mumol/L enzastaurin. Moreover, enzastaurin-induced cell death was not affected by interleukin-6 or ZVAD-fmk. GSK3beta phosphorylation, a reliable pharmacodynamic marker for enzastaurin activity, and AKT phosphorylation were both decreased with enzastaurin treatment. These data indicate that enzastaurin induces apoptosis in MM cell lines in a caspase-independent manner and that enzastaurin exerts its antimyeloma effect by inhibiting signaling through the AKT pathway.
Collapse
Affiliation(s)
- Mujahid A Rizvi
- Division of Hematology/Oncology, Department of Medicine, Northwestern University Feinberg School of Medicine, Lurie Building 3-250, 303 East Superior Street, Chicago, IL 60611, USA.
| | | | | | | | | | | | | | | |
Collapse
|
27
|
Abstract
Gliomas are the most common major subgroup of primary CNS tumours. Approximately 17,000 new cases are reported each year and, of these, 11,500 patients die. Glioblastoma multiforme (GBM) is highly proliferative and typically invades distal portions of the brain, thereby making complete surgical resection of these tumours nearly impossible. Moreover, GBMs are often resistant to current chemotherapy and radiation regimens. Therefore, there is a need for better therapeutic interventions. One class of proteins that is involved in the formation of malignant brain tumours is protein kinase C (PKC) and these kinases have not been thoroughly explored for their chemotherapeutic value in GBMs. The PKC isozyme, PKCeta (PKC-eta) increases cell proliferation and resistance to radiation of GBM cell lines. These properties make PKCeta an attractive target for chemotherapeutic intervention in the management of GBMs.
Collapse
Affiliation(s)
- Patrick M Martin
- Department of Pathology, University of Virginia, Charlottesville, VA, USA.
| | | |
Collapse
|
28
|
Wang Q, Wang X, Zhou Y, Evers BM. PKCdelta-mediated regulation of FLIP expression in human colon cancer cells. Int J Cancer 2006; 118:326-34. [PMID: 16052516 PMCID: PMC1850992 DOI: 10.1002/ijc.21373] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
FLICE-like inhibitory protein (FLIP), a naturally occurring caspase-inhibitory protein that lacks the critical cysteine domain necessary for catalytic activity, is a negative regulator of Fas-induced apoptosis. Decreased FLIP levels sensitize tumor cells to Fas- and TRAIL-mediated apoptosis; however, the cellular mechanisms regulating FLIP expression have not been defined. Here, we examined the roles of the PKC and NF-kappaB pathway in the regulation of FLIP in human colon cancers. FLIP mRNA levels were increased in Caco-2 cells by treatment with PMA; actinomycin D completely inhibited the induction of FLIP by PMA, indicating transcriptional regulation. PKC inhibitors Gö6983 and Ro-31-8220 blocked PMA-stimulated FLIP expression. Pretreatment with the PKCdelta-selective inhibitor rottlerin or transfection with PKCdelta siRNA inhibited PMA-induced FLIP expression, which identifies a role for PKCdelta in FLIP induction. Treatment with the proteasome inhibitor, MG132, or the NF-kappaB inhibitor (e.g., PDTC and gliotoxin), or overexpression of the superrepressor of IkappaB-alpha inhibited PMA-induced upregulation of FLIP. Moreover, PMA-induced NF-kappaB transactivation was blocked by GF109203x. In conclusion, our results demonstrate a critical role for PKCdelta/NF-kappaB in the regulation of FLIP in human colon cancer cells.
Collapse
Affiliation(s)
- Qingding Wang
- Department of Surgery, University of Texas Medical Branch, Galveston, TX 77555, USA
| | | | | | | |
Collapse
|
29
|
Liao YF, Hung YC, Chang WH, Tsay GJ, Hour TC, Hung HC, Liu GY. The PKC delta inhibitor, rottlerin, induces apoptosis of haematopoietic cell lines through mitochondrial membrane depolarization and caspases' cascade. Life Sci 2005; 77:707-19. [PMID: 15922001 DOI: 10.1016/j.lfs.2005.01.010] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/24/2004] [Accepted: 01/11/2005] [Indexed: 10/25/2022]
Abstract
Rottlerin is a widely selective protein kinase C delta (PKCdelta) inhibitor isolated from Mallotus philippinensis. It shown to be effective against several human tumor cell lines and in potentiating chemotherapy-induced cytotoxcicity. Using the trypan blue exclusion assay, we demonstrated that rottlerin reduced the viability in a dose- and time-dependent manner of human leukemia HL60 cells, human acute T cell leukemia Jurkat cells and mouse macrophage RAW 264.7 cells. Rottlerin caused apoptosis and the apaptotic processing was inhibited by a caspase inhibitor, z-VAD-fmk, in these haematopoietic cells. The apoptosis-inducing activities were determined by nuclear condensation, sub-G1 appearance, DNA fragmentation, loss of mitochondrial membrane potential (Deltapsim), release of mitochondrial cytochrome c into cytoplasm and proteolytic activation of caspase 9 and 3. Expression of PKCdelta and Bcl-2 protein inhibited Deltapsim change and repressed cell death. These studies suggest that the cytotoxic effects of rottlerin through inhibition of PKCdelta cause mitochondrial dysfunction, cytochrome c release from mitochondria into cytoplasm and the activation of caspases' cascade.
Collapse
Affiliation(s)
- Ya-Fan Liao
- Department of Life Sciences, National Chung-Hsing University, Taichung, Taiwan, ROC
| | | | | | | | | | | | | |
Collapse
|
30
|
Jelacic T, Linnekin D. PKCδ plays opposite roles in growth mediated by wild-type Kit and an oncogenic Kit mutant. Blood 2005; 105:1923-9. [PMID: 15542581 DOI: 10.1182/blood-2004-04-1450] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractThe Kit receptor tyrosine kinase is critical for normal hematopoiesis. Mutation of the aspartic acid residue encoded by codon 816 of human c-kit or codon 814 of the murine gene results in an oncogenic form of Kit. Here we investigate the role of protein kinase Cδ (PKCδ) in responses mediated by wild-type murine Kit and the D814Y mutant in a murine mast cell-like line. PKCδ is activated after wild-type (WT) Kit binds stem cell factor (SCF), is constitutively active in cells expressing the Kit catalytic domain mutant, and coprecipitates with both forms of Kit. Inhibition of PKCδ had opposite effects on growth mediated by wild-type and mutant Kit. Both rottlerin and a dominant-negative PKCδ construct inhibited the growth of cells expressing mutant Kit, while SCF-induced growth of cells expressing wild-type Kit was not inhibited. Further, overexpression of PKCδ inhibited growth of cells expressing wild-type Kit and enhanced growth of cells expressing the Kit mutant. These data demonstrate that PKCδ contributes to factor-independent growth of cells expressing the D814Y mutant, but negatively regulates SCF-induced growth of cells expressing wild-type Kit. This is the first demonstration that PKCδ has different functions in cells expressing normal versus oncogenic forms of a receptor.
Collapse
Affiliation(s)
- Tanya Jelacic
- Basic Research Laboratory, Center for Cancer Research, Bldg 469, Rm 205, National Cancer Institute-Frederick, Frederick, MD 21702, USA.
| | | |
Collapse
|
31
|
Pettersson F, Couture MC, Hanna N, Miller WH. Enhanced retinoid-induced apoptosis of MDA-MB-231 breast cancer cells by PKC inhibitors involves activation of ERK. Oncogene 2004; 23:7053-66. [PMID: 15273718 DOI: 10.1038/sj.onc.1207956] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
Retinoids are vitamin A derivatives, which cause growth inhibition, differentiation and/or apoptosis in various cell types, including some breast cancer cells. In general, estrogen receptor (ER)-positive cells are retinoic acid (RA) sensitive, whereas ER-negative cells are resistant. In this report, we show that ER-negative MDA-MB-231 cells are strongly growth inhibited by retinoids in combination with a PKC inhibitor. While neither RA nor GF109203X (GF) has a significant growth inhibitory effect in these cells, RA+GF potently suppress proliferation. We found that RA+GF induce apoptosis, as shown by an increase in fragmented DNA, Annexin-V-positive cells and caspase-3 activation. Apoptosis was also induced by GF in combination with two synthetic retinoids. Expression of phosphorylated as well as total PKC was decreased by GF and this was potentiated by RA. In addition, treatment with GF caused a strong and sustained activation of ERK1/2 and p38-MAPK, as well as a weaker activation of JNK. Importantly, inhibition of ERK but not p38 or JNK suppressed apoptosis induced by RA+GF, indicating that activation of ERK is specifically required. In support of this novel finding, the ability of other PKC inhibitors to cause apoptosis in combination with RA correlates with ability to cause sustained activation of ERK.
Collapse
Affiliation(s)
- Filippa Pettersson
- Lady Davis Institute for Medical Research, McGill University, 3755 Cote-Ste-Catherine Rd, Montreal, Quebec, Canada H3T 1E2
| | | | | | | |
Collapse
|
32
|
Jackson DN, Foster DA. The enigmatic protein kinase Cdelta: complex roles in cell proliferation and survival. FASEB J 2004; 18:627-36. [PMID: 15054085 DOI: 10.1096/fj.03-0979rev] [Citation(s) in RCA: 159] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Protein kinase Cdelta (PKCdelta) has been implicated both as a tumor suppressor and a positive regulator of cell cycle progression. PKCdelta has also been reported to positively and negatively regulate apoptotic programs. This has led to conflicting hypotheses on the role of PKCdelta in the control of cell proliferation and survival. Surprisingly, PKCdelta mice develop normally and are fertile, indicating that PKCdelta is not critical for normal cell proliferation during development. However, PKCdelta may play important roles in neoplastic cell proliferation. In this review, we have summarized the apparent multifunctional properties of this enigmatic protein with regard to its role in the regulation of cell cycle progression and cell survival. It is proposed that PKCdelta has both tumor suppressor and proliferation capabilities that can be recruited as a backup kinase for both gatekeeper tumor suppression and as an activator of the Ras/Raf/MEK/MAP kinase signaling pathway in cell proliferation.
Collapse
Affiliation(s)
- Desmond N Jackson
- Department of Biological Sciences, Hunter College of The City University of New York, New York, NY 10021, USA
| | | |
Collapse
|
33
|
Khan SB, Maududi T, Barton K, Ayers J, Alkan S. Analysis of histone deacetylase inhibitor, depsipeptide (FR901228), effect on multiple myeloma. Br J Haematol 2004; 125:156-61. [PMID: 15059137 DOI: 10.1111/j.1365-2141.2004.04882.x] [Citation(s) in RCA: 81] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Multiple myeloma (MM) is a neoplastic proliferation of plasma cells and remains an incurable disease because of the development of drug resistance. Histone deacytylase (HDAC) inhibitors are a new class of chemotherapeutic reagents that cause growth arrest and apoptosis of neoplastic cells. Depsipeptide, a new member of the HDAC inhibitors, was found to be safe in humans and has been shown to induce apoptosis in various cancers. In order to evaluate the effects of depsipeptide, a MM cell line, U266 [interleukin (IL)-6 dependent], was analysed for viability and apoptosis. The combined effect of depsipeptide with melphalan and changes in BCL-2 family proteins (BCL-2, BCL-XL, BAX and MCL-1) were also investigated. In addition, the RPMI 8226 cell line (IL-6 independent), and primary patient myeloma cells were also analysed for apoptosis after depsipeptide treatment. Depsipeptide induced apoptosis in both U266 and RPMI 8226 cell lines in a time- and dose-dependent fashion, and in primary patient myeloma cells. We also demonstrated that depsipeptide had an additive effect with melphalan (10 micromol/l). BCL-2, BCL-XL and MCL-1 showed decreased expression in depsipeptide-treated samples. Based on recent clinical trials demonstrating minimal clinical toxicity, our study supports the future clinical utilization of depsipeptide in the management of MM.
Collapse
Affiliation(s)
- S B Khan
- Department of Pathology, Loyola University Medical Center, 21660 South First Avenue, Maywood, IL 60153, USA
| | | | | | | | | |
Collapse
|