1
|
Li W, Zhu K, Liu Y, Liu M, Chen Q. Recent advances in PKC inhibitor development: Structural design strategies and therapeutic applications. Eur J Med Chem 2025; 287:117290. [PMID: 39904144 DOI: 10.1016/j.ejmech.2025.117290] [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: 12/16/2024] [Revised: 01/09/2025] [Accepted: 01/15/2025] [Indexed: 02/06/2025]
Abstract
Protein kinase C (PKC) isozymes play critical roles in diverse cellular processes and are implicated in numerous diseases, including cancer, diabetes, and autoimmune disorders. Despite extensive research efforts spanning four decades, only one PKC inhibitor has received clinical approval, highlighting the challenges in developing selective and efficacious PKC-targeting therapeutics. Here we review recent advances in the development of small-molecule PKC inhibitors, focusing on structural design strategies, pharmacological activities, and structure-activity relationships. We analyze emerging approaches including fragment-based drug design, allosteric targeting, and natural product derivatization that have yielded promising new scaffold classes. Special attention is given to innovations in achieving isozyme selectivity, particularly for PKCα and PKCβ, which have proven crucial for therapeutic applications. We discuss how integration of computational methods, structural biology insights, and rational design principles has advanced our understanding of PKC inhibition mechanisms. This comprehensive analysis reveals key challenges in PKC drug development, including the need for enhanced selectivity and reduced off-target effects, while highlighting promising directions for future therapeutic development. Our findings provide a framework for designing next-generation PKC inhibitors with improved clinical potential.
Collapse
Affiliation(s)
- Wen Li
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Kun Zhu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Yuyin Liu
- Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China
| | - Meixi Liu
- Department of Endocrinology, Deyang Hospital Affiliated Hospital of Chengdu University of Traditional Chinese Medicine, Deyang, 618000, China
| | - Qiu Chen
- Department of Endocrinology, Hospital of Chengdu University of Traditional Chinese Medicine, Chengdu, 610072, China.
| |
Collapse
|
2
|
Zheng H, Wang H, Zhang D, Gan Y, Wu Y, Xiang W, Fu P. Identification of therapeutic targets and immune landscape in glioblastoma through crosstalk with glioma-associated mesenchymal stem cells. Int Immunopharmacol 2025; 150:114228. [PMID: 39946771 DOI: 10.1016/j.intimp.2025.114228] [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: 11/16/2024] [Revised: 01/18/2025] [Accepted: 02/02/2025] [Indexed: 03/03/2025]
Abstract
BACKGROUND Glioma-associated mesenchymal stem cells (GA-MSCs) are one of the key factors limiting the effectiveness of glioblastoma (GBM) treatment and contributing to poor patient prognosis, making them a potential therapeutic target for GBM. In-depth research into the complex crosstalk between GA-MSCs and GBM cells not only aids in understanding the mechanisms of GBM progression but also provides valuable insights for developing new potential drugs. METHODS We conducted a comprehensive bioinformatics analysis aimed at identifying shared dysregulated genes between GBM and GA-MSCs. Through hub gene enrichment and immune infiltration analyses, we explored key molecular pathways and the immune landscape. Additionally, Cox regression analysis was employed to identify key factors influencing overall survival in GBM. The expression patterns and functional roles of hub genes were validated across various cancer types and datasets. Finally, dynamic simulations were used to assess the binding affinity of potential drugs to the targets, further supporting their potential as therapeutic candidates. RESULTS We identified 32 candidate genes primarily involved in the 1-kappa-B kinase/NF-kappa-B and MAPK signaling pathways, both of which played critical roles in tumor survival, proliferation, and invasion. Notable hub genes included DUSP1, FYN, FLNC, FN1, G3BP1, MYO1B, and WLS, each contributing uniquely to GBM progression. Among them, FLNC was highlighted as a key regulatory factor in GBM progression. Molecular dynamics simulations further revealed its potential as a therapeutic target, particularly demonstrating a high binding affinity with staurosporine. Additionally, a high proportion of dendritic cells contributed to the formation of the GBM immune microenvironment. CONCLUSIONS This study revealed the co-expression patterns and metabolic pathways between GA-MSCs and GBM, providing new insights into the molecular mechanisms of GBM progression. Targeting FLNC with staurosporine presents a promising therapeutic strategy for GBM. Aditionally, targeting the shared pathways of both may offer a valuable approach for treating malignant brain tumors.
Collapse
Affiliation(s)
- Haoyang Zheng
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| | - Haofei Wang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| | - Duo Zhang
- Department of Nursing, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030 China
| | - Yong Gan
- Department of Social Medicine and Health Management, School of Public Health, Tongji Medical College, Huazhong University of Science and Technology, No. 13 Hangkong Road, Wuhan 430030 China
| | - Yuyi Wu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China
| | - Wei Xiang
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China.
| | - Peng Fu
- Department of Neurosurgery, Union Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430022 China.
| |
Collapse
|
3
|
Pelargonidin inhibits vascularization and metastasis of brain gliomas by blocking the PI3K/AKT/mTOR pathway. J Biosci 2022. [DOI: 10.1007/s12038-022-00281-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
|
4
|
Trendowski M. Exploiting the cytoskeletal filaments of neoplastic cells to potentiate a novel therapeutic approach. Biochim Biophys Acta Rev Cancer 2014; 1846:599-616. [PMID: 25286320 DOI: 10.1016/j.bbcan.2014.09.007] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/02/2014] [Revised: 09/19/2014] [Accepted: 09/21/2014] [Indexed: 02/06/2023]
Abstract
Although cytoskeletal-directed agents have been a mainstay in chemotherapeutic protocols due to their ability to readily interfere with the rapid mitotic progression of neoplastic cells, they are all microtubule-based drugs, and there has yet to be any microfilament- or intermediate filament-directed agents approved for clinical use. There are many inherent differences between the cytoskeletal networks of malignant and normal cells, providing an ideal target to attain preferential damage. Further, numerous microfilament-directed agents, and an intermediate filament-directed agent of particular interest (withaferin A) have demonstrated in vitro and in vivo efficacy, suggesting that cytoskeletal filaments may be exploited to supplement chemotherapeutic approaches currently used in the clinical setting. Therefore, this review is intended to expose academics and clinicians to the tremendous variety of cytoskeletal filament-directed agents that are currently available for further chemotherapeutic evaluation. The mechanisms by which microfilament directed- and intermediate filament-directed agents damage malignant cells are discussed in detail in order to establish how the drugs can be used in combination with each other, or with currently approved chemotherapeutic agents to generate a substantial synergistic attack, potentially establishing a new paradigm of chemotherapeutic agents.
Collapse
Affiliation(s)
- Matthew Trendowski
- Department of Biology, Syracuse University, 107 College Place, Syracuse, NY 13244, USA.
| |
Collapse
|
5
|
Mukthavaram R, Jiang P, Saklecha R, Simberg D, Bharati IS, Nomura N, Chao Y, Pastorino S, Pingle SC, Fogal V, Wrasidlo W, Makale M, Kesari S. High-efficiency liposomal encapsulation of a tyrosine kinase inhibitor leads to improved in vivo toxicity and tumor response profile. Int J Nanomedicine 2013; 8:3991-4006. [PMID: 24174874 PMCID: PMC3808212 DOI: 10.2147/ijn.s51949] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Staurosporine (STS) is a potent pan-kinase inhibitor with marked activity against several chemotherapy-resistant tumor types in vitro. The translational progress of this compound has been hindered by poor pharmacokinetics and toxicity. We sought to determine whether liposomal encapsulation of STS would enhance antitumor efficacy and reduce toxicity, thereby supporting the feasibility of further preclinical development. We developed a novel reverse pH gradient liposomal loading method for STS, with an optimal buffer type and drug-to-lipid ratio. Our approach produced 70% loading efficiency with good retention, and we provide, for the first time, an assessment of the in vivo antitumor activity of STS. A low intravenous dose (0.8 mg/kg) inhibited U87 tumors in a murine flank model. Biodistribution showed preferential tumor accumulation, and body weight data, a sensitive index of STS toxicity, was unaffected by liposomal STS, but did decline with the free compound. In vitro experiments revealed that liposomal STS blocked Akt phosphorylation, induced poly(ADP-ribose) polymerase cleavage, and produced cell death via apoptosis. This study provides a basis to explore further the feasibility of liposomally encapsulated STS, and potentially related compounds for the management of resistant solid tumors.
Collapse
Affiliation(s)
- Rajesh Mukthavaram
- Translational Neuro-Oncology Laboratories, Moores Cancer Center, UC San Diego, La Jolla, CA, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Targeting sphingosine kinase 1 in carcinoma cells decreases proliferation and survival by compromising PKC activity and cytokinesis. PLoS One 2012; 7:e39209. [PMID: 22761740 PMCID: PMC3382615 DOI: 10.1371/journal.pone.0039209] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/02/2012] [Accepted: 05/21/2012] [Indexed: 11/20/2022] Open
Abstract
Sphingosine kinases (SK) catalyze the phosphorylation of proapoptotic sphingosine to the prosurvival factor sphingosine 1-phosphate (S1P), thereby promoting oncogenic processes. Breast (MDA-MB-231), lung (NCI-H358), and colon (HCT 116) carcinoma cells were transduced with shRNA to downregulate SK-1 expression or treated with a pharmacologic SK-1 inhibitor. The effects of SK-1 targeting were investigated by measuring the level of intracellular sphingosine, the activity of protein kinase C (PKC) and cell cycle regulators, and the mitotic index. Functional assays included measurement of cell proliferation, colony formation, apoptosis, and cell cycle analysis. Downregulation of SK-1 or its pharmacologic inhibition increased intracellular sphingosine and decreased PKC activity as shown by reduced phosphorylation of PKC substrates. In MDA-MB-231 cells this effect was most pronounced and reduced cell proliferation and colony formation, which could be mimicked using exogenous sphingosine or the PKC inhibitor RO 31-8220. SK-1 downregulation in MDA-MB-231 cells increased the number of cells with 4N and 8N DNA content, and similar effects were observed upon treatment with sphingosine or inhibitors of SK-1 or PKC. Examination of cell cycle regulators unveiled decreased cdc2 activity and expression of Chk1, which may compromise spindle checkpoint function and cytokinesis. Indeed, SK-1 kd cells entered mitosis but failed to divide, and in the presence of taxol also failed to sustain mitotic arrest, resulting in further increased endoreduplication and apoptosis. Our findings delineate an intriguing link between SK-1, PKC and components of the cell cycle machinery, which underlines the significance of SK-1 as a target for cancer therapy.
Collapse
|
7
|
Ye CP, Yano S, Tfelt-Hansen J, MacLeod RJ, Ren X, Terwilliger E, Brown EM, Chattopadhyay N. Regulation of a Ca2+-activated K+ channel by calcium-sensing receptor involves p38 MAP kinase. J Neurosci Res 2004; 75:491-8. [PMID: 14743432 DOI: 10.1002/jnr.10875] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
By using pharmacological and molecular approaches, we previously showed that the G-protein-coupled, extracellular calcium (Ca2+(o))-sensing receptor (CaR) regulates a large-conductance (approximately 140 pS), Ca(2+)-activated K+ channel [IK(Ca); CAKC] in U87 astrocytoma cells. Here we show that elevated Ca2+(o) stimulates extracellular-signal-regulated kinase (ERK1/2) and p38 MAP kinase (MAPK). The effect of high Ca2+(o) on p38 MAPK but not ERK1/2 is CaR mediated, insofar as transduction with a dominant-negative CaR (R185Q) using recombinant adeno-associated virus (rAAV) attenuated the activation of p38 MAPK but not of ERK1/2. p38 MAPK activation by the CaR is likely to be protein kinase C (PKC) independent, in that the pan-PKC inhibitor GF109203X failed to abolish the high-Ca2+(o)-induced phosphorylation of p38 MAPK. Consistently with our data on the activation of this kinase, we observed that inhibiting p38 MAPK blocked the activation of the CAKC induced by the specific pharmacological CaR activator NPS R-467. In contrast, inhibiting MEK1 only transiently inhibited the activation of this K+ channel by NPS R-467, despite the continued presence of the antagonist. Similarly to the lack of any effect of the PKC inhibitor on the activation of ERK1/2 and p38 MAPK, inhibiting PKC had no effect on NPS R-467-induced activation of this channel. Therefore, our data show that the CaR, acting via p38 MAPK, regulates a large-conductance CAKC in U87 cells, a process that is PKC independent. Large-conductance CAKCs play an important role in the regulation of cellular volume, so our results have important implications for glioma cell volume regulation.
Collapse
Affiliation(s)
- Chian Ping Ye
- Division of Endocrinology, Hypertension and Diabetes and Membrane Biology Program, Department of Medicine, Brigham and Women's Hospital, Harvard Medical School, Boston, Massachusetts 02115, USA.
| | | | | | | | | | | | | | | |
Collapse
|
8
|
Yamasaki F, Hama S, Yoshioka H, Kajiwara Y, Yahara K, Sugiyama K, Heike Y, Arita K, Kurisu K. Staurosporine-induced apoptosis is independent of p16 and p21 and achieved via arrest at G2/M and at G1 in U251MG human glioma cell line. Cancer Chemother Pharmacol 2003; 51:271-83. [PMID: 12721754 DOI: 10.1007/s00280-002-0562-z] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2002] [Accepted: 11/08/2002] [Indexed: 11/27/2022]
Abstract
OBJECTIVE The mechanisms involved in the cell cycle and cell death remain unresolved despite much investigation. Staurosporine induces cell death and G1 or G2/M arrest in a dose-dependent manner, but the mechanisms remain unknown. METHODS In the present study an adenovirus vector expressing p16 or p21 genes in human glioma cell lines was used to examine cell cycle regulation and cell death induced by staurosporine. RESULTS A low concentration (</=10 n M) of staurosporine induced G1 arrest of U251MG cells, whereas a high concentration (>/=30 n M) induced G2/M arrest and finally induced apoptosis via a caspase-3-activated pathway from both the G2/M and G1 phases. However, pRb was dephosphorylated and cdc2 was inhibited at both the low and the high concentrations of staurosporine, indicating that the mechanisms of cell cycle regulation are not simply p53-Rb- or cdc2-dependent pathways. CONCLUSIONS Forced G1 arrest by transfection with p16 or p21 genes did not alter the rate of staurosporine-induced cell death. This implies that an unknown pathway of apoptosis occurs from the G1 phase.
Collapse
Affiliation(s)
- Fumiyuki Yamasaki
- Department of Neurosurgery, Faculty of Medicine, Hiroshima University, 1-2-3 Kasumi, Minami-ku, Hiroshima, Japan.
| | | | | | | | | | | | | | | | | |
Collapse
|
9
|
da Rocha AB, Mans DRA, Regner A, Schwartsmann G. Targeting protein kinase C: new therapeutic opportunities against high-grade malignant gliomas? Oncologist 2002; 7:17-33. [PMID: 11854544 DOI: 10.1634/theoncologist.7-1-17] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
A large body of evidence suggests that the abnormal phenotype of neoplastic astrocytes, including their excessive proliferation rate and high propensity to invade surrounding tissues, results from mutations in critical genes involved in key cellular events. These genetic alterations can affect cell-surface-associated receptors, elements of signaling pathways, or components of the cell cycle clock, conferring a gain or a loss of relevant metabolic functions of the cells. The understanding of such phenomena may allow the development of more efficacious forms of cancer treatment. Examples are therapies specifically directed against overexpressed epidermal growth factor receptor, hyperactive Ras, excessively stimulated Raf-1, overproduced ornithine decarboxylase, or aberrantly activated cyclin-dependent kinases. The applicability of some of these approaches is now being assessed in patients suffering from primary malignant central nervous system tumors that are not amenable to current therapeutic modalities. Another potentially useful therapeutic strategy against such tumors involves the inhibition of hyperactive or overexpressed protein kinase C (PKC). This strategy is justified by the decrease in cell proliferation and invasion following inhibition of the activity of this enzyme observed in preclinical glioma models. Thus, interference with PKC activity may represent a novel form of experimental cancer treatment that may simultaneously restrain the hyperproliferative state and the invasive capacity of high-grade malignant gliomas without inducing the expected toxicity of classical cytotoxic agents. Of note, the experimental use of PKC-inhibiting agents in patients with refractory high-grade malignant gliomas has indeed led to some clinical responses. The present paper reviews the current status of the biochemistry and molecular biology of PKC, as well as the possibilities for developing novel anti-PKC-based therapies for central nervous system malignancies.
Collapse
Affiliation(s)
- A B da Rocha
- South-American Office for Anticancer Drug Development (SOAD), Comprehensive Cancer Center, Lutheran University of Brazil, Canoas, RS, Brazil.
| | | | | | | |
Collapse
|
10
|
Jiang F, Cho KK, Mikkelse T, Tong L, Lew YS, Hochbaum N, Shargorodsky J, Chop M. Tamoxifen increases photodynamic therapeutic response of U87 and U25ln human glioma cells. J Neurooncol 2002; 56:51-8. [PMID: 11949827 DOI: 10.1023/a:1014400822235] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
We tested the hypothesis that Tamoxifen (TMX), an inhibitor of protein kinase C (PKC), augments the cytotoxicity of photodynamic therapy (PDT) treatment of human (U87) and (U25ln) glioma cells. U87 and U25ln glioma cells were plated and treated with PDT using Photofrin as the sensitizer. Cells were treated with Photofrin at various doses and with various optical (632 nm) irradiation intensities 24 h later. Cells were also treated with Photofrin at a fixed dose alone and with various doses of Tamoxifen and subjected to laser treatment 24 h later. Tumor response was tested using the (3-94,5-dimethyl-2-yl)-2,5-diphenyl-tetrazolium (MTT) method. Total toxicity of U87 cells was achieved with PDT at all doses of Photofrin (1, 2.5, 5, 10 microg/ml) with irradiation densities equal to or greater than 200 mJ/cm2. Using an irradiation intensity of 100 mJ/cm2, U87 and U25ln cells were killed in a Photofrin dose-dependent manner. Significant cytotoxicity was detected with Photofrin doses of 5 microg/ml (p < 0.05) and 10 microg/ml (p < 0.001). Tamoxifen at a dose of 500 microg/ml and higher, significantly increased the toxicity of the PDT response with 5 microg/ml Photofrin and 100 mJ/cm2 (p < 0.05). In summary, our data demonstrate that Tamoxifen significantly enhances the Photofrin PDT activity of U87 and U25ln human glioma cells.
Collapse
Affiliation(s)
- Feng Jiang
- Department of Neurology, Henry Ford Health Sciences, Detroit, MI, USA
| | | | | | | | | | | | | | | |
Collapse
|
11
|
Abstract
PKC isoenzymes were found to be involved in proliferation, antitumor drug resistance and apoptosis. Therefore, it has been tried to exploit PKC as a target for antitumor treatment. PKC alpha activity was found to be elevated, for example, in breast cancers and malignant gliomas, whereas it seems to be underexpressed in many colon cancers. So it can be expected that inhibition of PKC activity will not show similar antitumor activity in all tumors. In some tumors it seems to be essential to inhibit PKC to reduce growth. However, for inhibition of tumor proliferation it may be an advantage to induce apoptosis. In this case an activation of PKC delta should be achieved. The situation is complicated by the facts that bryostatin leads to the activation of PKC and later to a downmodulation and that the PKC inhibitors available to date are not specific for one PKC isoenzyme. For these reasons, PKC modulation led to many contradicting results. Despite these problems, PKC modulators such as miltefosine, bryostatin, safingol, CGP41251 and UCN-01 are used in the clinic or are in clinical evaluation. The question is whether PKC is the major or the only target of these compounds, because they also interfere with other targets. PKC may also be involved in apoptosis. Oncogenes and growth factors can induce cell proliferation and cell survival, however, they can also induce apoptosis, depending on the cell type or conditions in which the cells or grown. PKC participates in these signalling pathways and cross-talks. Induction of apoptosis is also dependent on many additional factors, such as p53, bcl-2, mdm2, etc. Therefore, there are also many contradicting results on PKC modulation of apoptosis. Similar controversial data have been reported about MDR1-mediated multidrug resistance. At present it seems that PKC inhibition alone without direct interaction with PGP will not lead to successful reversal of PGP-mediated drug efflux. One possibility to improve chemotherapy would be to combine established antitumor drugs with modulators of PKC. However, here also very contrasting results were obtained. Many indicate that inhibition, others, that activation of PKC enhances the antiproliferative activity of anticancer drugs. The problem is that the exact functions of the different PKC isoenzymes are not clear at present. So further investigations into the role of PKC isoenzymes in the complex and interacting signalling pathways are essential. It is a major challenge in the future to reveal whether modulation of PKC can be used for the improvement of cancer therapy.
Collapse
Affiliation(s)
- J Hofmann
- Institute of Medical Chemistry and Biochemistry, University of Innsbruck, A-6020 Innsbruck, Austria
| |
Collapse
|
12
|
Besson A, Yong VW. Involvement of p21(Waf1/Cip1) in protein kinase C alpha-induced cell cycle progression. Mol Cell Biol 2000; 20:4580-90. [PMID: 10848585 PMCID: PMC85854 DOI: 10.1128/mcb.20.13.4580-4590.2000] [Citation(s) in RCA: 98] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Protein kinase C (PKC) plays an important role in the regulation of glioma growth; however, the identity of the specific isoform and mechanism by which PKC fulfills this function remain unknown. In this study, we demonstrate that PKC activation in glioma cells increased their progression through the cell cycle. Of the six PKC isoforms that were present in glioma cells, PKC alpha was both necessary and sufficient to promote cell cycle progression when stimulated with phorbol 12-myristate 13-acetate. Also, decreased PKC alpha expression resulted in a marked decrease in cell proliferation. The only cell cycle-regulatory molecule whose expression was rapidly altered and increased by PKC alpha activity was the cyclin-cyclin-dependent kinase (CDK) inhibitor p21(Waf1/Cip1). Coimmunoprecipitation studies revealed that p21(Waf1/Cip1) upregulation was accompanied by an incorporation of p21(Waf1/Cip1) into various cyclin-CDK complexes and that the kinase activity of these complexes was increased, thus resulting in cell cycle progression. Furthermore, depletion of p21(Waf1/Cip1) by antisense strategy attenuated the PKC-induced cell cycle progression. These results suggest that PKC alpha activity controls glioma cell cycle progression through the upregulation of p21(Waf1/Cip1), which facilitates active cyclin-CDK complex formation.
Collapse
Affiliation(s)
- A Besson
- Departments of Oncology and Clinical Neurosciences, University of Calgary, Canada
| | | |
Collapse
|
13
|
Cho KK, Mikkelsen T, Lee YJ, Jiang F, Chopp M, Rosenblum ML. The role of protein kinase Calpha in U-87 glioma invasion. Int J Dev Neurosci 1999; 17:447-61. [PMID: 10571407 DOI: 10.1016/s0736-5748(99)00054-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/04/2023] Open
Abstract
To investigate the hypothesis that protein kinase Calpha (PKCalpha) is functional glial tumor cell invasion, stable PKCalpha sense and antisense transfected U-87 cell lines were established and PKCalpha expression characterized by Western blot and PKC activity assays. Invasion assays including barrier migration (Koochekpour et al., Extracellular matrix proteins inhibit proliferation, upregulate migration and induce morphological changes in human glioma lines. Eur. J. Cancer, 1995, 31, 375-380; Merzak et al., CD44 mediates human glioma cell adhesion and invasion in vitro. Cancer Res., 1994, 54, 3988-3992; Merzak et al., Cell surface gangliosides are involved in the control of human glioma cell invasion in vitro. Neurosci. Lett., 1994, 177, 11-16), and spheroid confrontation were used to study the relationship between PKCalpha expression and invasiveness. PKCalpha overexpressing clones show increased barrier migration (1.5x) relative to the control transfected clones. PKCalpha inhibited clones exhibited reduced invasiveness, to < 50%. In coculture with PKCalpha overexpressing clones, the remaining normal fetal rat brain aggregate volume was significantly decreased (up to 200%) but 90% of the initial brain volume was left in PKCalpha inhibited clone in the rat brain aggregate tumor spheroid confrontation. This effect was not associated with significant growth inhibition. We conclude that expression of PKCalpha in glioma-derived cell lines appears to be central to glioma invasion in vitro.
Collapse
Affiliation(s)
- K K Cho
- Department of Neurosurgery, Henry Ford Hospital, Detroit, MI 48202, USA
| | | | | | | | | | | |
Collapse
|
14
|
Bredel M, Pollack IF, Freund JM, Rusnak J, Lazo JS. Protein kinase C inhibition by UCN-01 induces apoptosis in human glioma cells in a time-dependent fashion. J Neurooncol 1999; 41:9-20. [PMID: 10222418 DOI: 10.1023/a:1006047025425] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Recent studies in our laboratory have shown that UCN-01 (7-hydroxystaurosporine), which is a derivative of the non-selective protein kinase inhibitor staurosporine that exhibits relative selectivity for protein kinase C (PKC), is a potent inhibitor of glioma growth in in vitro and in vivo models. This agent exhibits both cytotoxic and cytostatic effects, depending on the time period of drug exposure. In the present study, we examined whether UCN-01-induced cytotoxicity correlated with the induction of apoptosis, and characterized further the time course of this process as a prelude to application of UCN-01 in clinical trials. We first demonstrated that the cytotoxic effects of UCN-01 were associated with the induction of morphological features of apoptosis. Secondly, we identified electrophoretic features of apoptosis semiquantitatively at a series of time points using field inversion gel electrophoresis. These studies showed a peak in the induction of high-molecular-weight DNA fragmentation after 3-6 days of drug treatment. Thirdly, we measured the percentage of cells undergoing apoptosis at various time points using a terminal transferase-catalyzed in situ end-labeling technique, which confirmed a time- and concentration-dependent increase in apoptotic cell numbers. This correlated with a progressive decrease in the percentage of cells that were viable as assessed by trypan blue exclusion. Cell killing peaked within 2-4 days after beginning UCN-01 treatment, but continued at a lower level in the ensuing days. Taken together, these studies demonstrated that extended periods of exposure to UCN-01 are needed for optimal manifestation of cytotoxic effects against glioma cells, a factor that must be taken into consideration in the design of future clinical trials with this agent for malignant gliomas.
Collapse
Affiliation(s)
- M Bredel
- Department of Neurosurgery, University of Pittsburgh School of Medicine, Pennsylvania, USA
| | | | | | | | | |
Collapse
|
15
|
Bredel M, Pollack IF. The role of protein kinase C (PKC) in the evolution and proliferation of malignant gliomas, and the application of PKC inhibition as a novel approach to anti-glioma therapy. Acta Neurochir (Wien) 1998; 139:1000-13. [PMID: 9442212 DOI: 10.1007/bf01411552] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
The present article reviews the role of the second messenger enzyme protein kinase C (PKC) in the growth regulation of high-grade gliomas, and evaluates the efficacy of therapeutic strategies directed against PKC for blocking the proliferation of these malignancies in in vitro and in vivo models. The translation of such strategies to the treatment of patients with malignant gliomas may provide a novel approach for improving the otherwise grim outlook associated with these neoplasms.
Collapse
Affiliation(s)
- M Bredel
- Department of Neurosurgery, University of Pittsburgh School of Medicine, PA, USA
| | | |
Collapse
|
16
|
Uhm JH, Dooley NP, Villemure JG, Yong VW. Mechanisms of glioma invasion: role of matrix-metalloproteinases. Neurol Sci 1997; 24:3-15. [PMID: 9043741 DOI: 10.1017/s0317167100021028] [Citation(s) in RCA: 87] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
One of the most lethal properties of high grade gliomas is their ability to invade the surrounding normal brain tissue, as infiltrated cells often escape surgical resection and inevitably lead to tumour recurrence. The consequent poor prognosis and survival rate underscore the need to further understand and target the cellular mechanisms that underly tumour invasiveness. Proteases which degrade the surrounding stromal cells and extracellular matrix proteins have been demonstrated to be critical effectors of invasion for tumours of both central and peripheral origin. Within the nervous system, the role of metalloproteinases as well as other classes of proteases in mediating the invasive phenotype of high grade gliomas has been an intense area of research. We present in this article a review of this literature and address the possibility that these proteases and the biochemical pathways that regulate their expression, such as protein kinase C, may represent potential targets in the therapy of high grade gliomas.
Collapse
Affiliation(s)
- J H Uhm
- Department of Neurology & Neurosurgery, Montreal Neurological Institute, McGill University, Canada
| | | | | | | |
Collapse
|
17
|
Baltuch GH, Yong VW. Signal transduction for proliferation of glioma cells in vitro occurs predominantly through a protein kinase C-mediated pathway. Brain Res 1996; 710:143-9. [PMID: 8963653 DOI: 10.1016/0006-8993(95)01395-4] [Citation(s) in RCA: 30] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Previous work has demonstrated that glioma cells have very high protein kinase C (PKC) enzyme activity when compared to non-malignant glia, and that their PKC activity correlates with their proliferation rate. The purpose of this study was to determine whether the elevated PKC activity in glioma is secondary to an autonomously active PKC isoform implying oncogenic transformation, or whether this activity is driven by upstream ligand-receptor tyrosine kinase interactions. We treated established human glioma cell lines A172, U563 or U251 with either the highly selective PKC inhibitor CGP 41 251, or with genistein, a tyrosine kinase inhibitor. The proliferation rate and PKC activity of all the glioma lines was reduced by CGP 41 251; the IC50 values for inhibiting cell proliferation corresponded to the IC50v values for inhibition of PKC activity. Genistein also inhibited cell proliferation, with IC50 proliferation values approximating those for inhibition of tyrosine kinase activity in cell free protein extracts. Importantly, in genistein-treated cells, downstream PKC enzyme activity was dose dependently reduced such that the correlation coefficient for effects of genistein on proliferation rate and PKC activity was 0.92. These findings suggest that upstream tyrosine kinase linked events, rather than an autonomously functioning PKC, result in the high PKC activity observed in glioma. Finally, fetal calf serum (FCS) evoked a strong mitogenic effect on glioma cell lines. This mitogenic activity was completely blocked by CGP 41 251, suggesting that although the many mitogens in FCS for glioma cells signal initially through genistein-inhibitable tyrosine kinases, they ultimately channel through a PKC-dependent pathway. We conclude that proliferative signal transduction in glioma cells occurs through a predominantly PKC-dependent pathway and that selectively targeting this enzyme provides an approach to glioma therapy.
Collapse
Affiliation(s)
- G H Baltuch
- Department of Neurology and Neurosurgery, Montreal Neurological Institute, McGill University, Quebec, Canada
| | | |
Collapse
|
18
|
Baltuch GH, Dooley NP, Villemure JG, Yong VW. Protein kinase C and growth regulation of malignant gliomas. Neurol Sci 1995; 22:264-71. [PMID: 8599768 DOI: 10.1017/s0317167100039457] [Citation(s) in RCA: 23] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
This article reviews the role of the signal transduction enzyme protein kinase C in the regulation of growth of malignant gliomas, and describes how targetting this enzyme clinically can provide a novel approach to glioma therapy.
Collapse
Affiliation(s)
- G H Baltuch
- Montreal Neurological Institute, Department of Neurology and Neurosurgery, McGill University, Quebec, Canada
| | | | | | | |
Collapse
|
19
|
Baltuch GH, Dooley NP, Rostworowski KM, Villemure JG, Yong VW. Protein kinase C isoform alpha overexpression in C6 glioma cells and its role in cell proliferation. J Neurooncol 1995; 24:241-50. [PMID: 7595754 DOI: 10.1007/bf01052840] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023]
Abstract
Previous studies from this laboratory have demonstrated that protein kinase C (PKC) enzyme activity is highly correlated with the proliferation rate of glioma cells, and that glioma cells of both human and rat origin have very high PKC enzyme activity when compared to non-malignant glia including astrocytes, the antecedents of most gliomas. In the present study, by contrasting the rat C6 glioma cells with non-malignant rat astrocytes, we have sought to determine whether the high PKC enzyme activity of glioma cells was due to the overexpression of a specific isoform of PKC. By Western blot analyses, both C6 glioma cells and astrocytes were found to express PKC alpha, beta, delta, epsilon and zeta, but not gamma. Enzyme activity measurements revealed that the elevated PKC activity of glioma cells compared to glia was calcium-dependent, thereby implicating abnormal activity of the alpha or beta isoforms. On Western blots, when compared to astrocytes, glioma cells were determined to overexpress PKC alpha but not beta. An antisense oligonucleotide to PKC alpha, directed at the site of initiation of translation, inhibited the proliferation rate of glioma cells when compared to cells treated with control oligonucleotides; PKC enzyme activity and PKC alpha protein expression were significantly reduced by the antisense treatment. These results suggest that the high PKC enzyme activity of glioma cells, and its correspondence with proliferation rate, is the result of overexpression of isozyme alpha. Targetting PKC alpha in glioma cells may provide a refinement of therapy of glioma patients, some of which are already showing clinical stabilization when treated with drugs with PKC-inhibitory effects.
Collapse
Affiliation(s)
- G H Baltuch
- Department of Neurology and Neurosurgery, Montreal Neurological Institute and McGill University, Quebec, Canada
| | | | | | | | | |
Collapse
|