1
|
Kou YY, Liu J, Chang YT, Liu LY, Sun F, Li YL, Leng JR, Lin HW, Yang F. Marine derived macrolide bryostatin 4 inhibits the TGF-β signaling pathway against acute erythroleukemia. Cell Oncol (Dordr) 2024; 47:1863-1878. [PMID: 39083211 DOI: 10.1007/s13402-024-00968-0] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 06/18/2024] [Indexed: 10/11/2024] Open
Abstract
PURPOSE Acute erythroleukemia (AEL) is a rare and highly aggressive subtype of acute myeloid leukemia (AML) with an extremely poor prognosis when treated with available drugs. Therefore, new investigational agents capable of inducing remission are urgently required. METHODS Bioinformatics analysis, western blot and qRT-PCR were used to reveal the potential biological mechanism of bryostatin 4 (B4), an antineoplastic macrolide derived from the marine bryozoan Bugula neritina. Then, in vivo experiments were conducted to evaluate the role of transforming growth factor (TGF)-β signaling in the progression of AEL. RESULTS Our results revealed that the proliferation of K562 cells and TF-1 cells was significantly inhibited by B4 at IC50 values of 37 nM and 52 nM, respectively. B4 inhibited TGF-β signaling and its downstream pathway targets, particularly the phosphorylation of Smad2, Smad3, Ras, C-RAF, ERK1/2, and MEK. B4 also played an important role in cell invasion and migration in K562 cells and TF-1 cells by reducing the protein levels of the mesenchymal cell marker vimentin. Moreover, Flow cytometry and western blot analyses demonstrated that B4 induced apoptosis and initiated G0/G1 phase arrest by modulating mitochondrial dysfunction and cyclin-dependent kinase (CDK) expression. CONCLUSION These findings indicated that B4 could inhibit the proliferation, migration, invasion, and TGF-β signaling pathways of AEL cells, thus suggesting that B4 possesses therapeutic potential as a treatment for AEL.
Collapse
Affiliation(s)
- Yan-Yu Kou
- Department of Pharmacy, Research Center for Marine Drugs, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200127, China
- School of Pharmacy, Shanghai JiaoTong University, Shanghai, China
| | - Jie Liu
- Department of Pharmacy, Research Center for Marine Drugs, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200127, China
| | - Yung-Ting Chang
- Department of Pharmacy, Research Center for Marine Drugs, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200127, China
| | - Li-Yun Liu
- Department of Pharmacy, Research Center for Marine Drugs, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200127, China
| | - Fan Sun
- Department of Pharmacy, Research Center for Marine Drugs, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200127, China
| | - Yi-Lin Li
- National Facility for Protein Science in Shanghai, Zhangjiang Lab, Shanghai Advanced Research Institute, Chinese Academy of Science, Shanghai, 201210, China
| | - Jia-Rong Leng
- Department of Pharmacy, Research Center for Marine Drugs, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200127, China
| | - Hou-Wen Lin
- Department of Pharmacy, Research Center for Marine Drugs, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200127, China.
| | - Fan Yang
- Department of Pharmacy, Research Center for Marine Drugs, Renji Hospital, School of Medicine, Shanghai JiaoTong University, Shanghai, 200127, China.
| |
Collapse
|
2
|
Fleming AK, Storz P. Protein kinase C isoforms in the normal pancreas and in pancreatic disease. Cell Signal 2017; 40:1-9. [PMID: 28826907 DOI: 10.1016/j.cellsig.2017.08.005] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2017] [Accepted: 08/17/2017] [Indexed: 12/16/2022]
Abstract
Protein Kinase C isoforms have been implicated in regulating multiple processes within the healthy pancreas. Moreover, their dysregulation contributes to all aspects of pancreatic disease. In this review, with a focus on acinar, ductal, and islet cells, we highlight the roles and contributions of the different PKC isoforms to normal pancreas function. We also discuss the contribution of PKC enzymes to pancreatic diseases, including insulin resistance and diabetes mellitus, as well as pancreatitis and the development and progression of pancreatic cancer.
Collapse
Affiliation(s)
- Alicia K Fleming
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA
| | - Peter Storz
- Department of Cancer Biology, Mayo Clinic, Jacksonville, FL, USA.
| |
Collapse
|
3
|
Gao X, Xu F, Zhang HT, Chen M, Huang W, Zhang Q, Zeng Q, Liu L. PKCα-GSK3β-NF-κB signaling pathway and the possible involvement of TRIM21 in TRAIL-induced apoptosis. Biochem Cell Biol 2016; 94:256-64. [PMID: 27219672 DOI: 10.1139/bcb-2016-0009] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023] Open
Abstract
Tumor necrosis factor related apoptosis-inducing ligand (TRAIL) is a highly promising therapeutic agent for cancer treatment, owing to its ability to selectively target tumor cells for cell death while having little effect on most normal cells. However, recent research has found that many cancers, including non-small cell lung cancer (NSCLC), display resistance to TRAIL. Therefore, it is important to elucidate the molecular mechanisms governing the resistance of tumor cells to TRAIL treatment. In this study, we show that GSK3β antagonized TRAIL-induced apoptosis in H1299 NSCLC cells, and determined that the PKCα isozyme is an upstream regulator of GSK3β that phosphorylates and inactivates GSK3β, thereby sensitizing cancer cells to TRAIL-induced apoptosis. Furthermore, we demonstrated that the anti-apoptotic effect of GSK3β is mediated by the NF-κB pathway, whereas the tripartite motif 21 (TRIM21) was able to inhibit the activation of NF-κB by GSK3β, and leads to the promotion of cell apoptosis. Taken together, our study further delineated the underpinning mechanism of resistance to TRAIL-induced apoptosis in H1299 cells, and provided new clues for sensitizing NSCLC cells to TRAIL therapy.
Collapse
Affiliation(s)
- Xuejuan Gao
- a Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, Jinan University, Guangzhou 510632, China
| | - Fengmei Xu
- a Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, Jinan University, Guangzhou 510632, China
| | - Huan-Tian Zhang
- b Institute of Orthopedic Diseases and Department of Orthopedics, the First Affiliated Hospital, Jinan University, Guangzhou 510630, China
| | - Miaojuan Chen
- c Department of Interventional Radiology and Vascular Anomalies, Guangzhou Women and Children's Medical Center, Guangzhou 510623, China
| | - Wensi Huang
- a Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, Jinan University, Guangzhou 510632, China
| | - Qihao Zhang
- d Institute of Biomedicine, and National Engineering Research Center of Genetic Medicine; Guangdong Provincial Key Laboratory of Bioengineering Medicine, Jinan University, Guangzhou 510632, China
| | - Qingzhong Zeng
- a Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, Jinan University, Guangzhou 510632, China
| | - Langxia Liu
- a Key Laboratory of Functional Protein Research of Guangdong Higher Education Institutes, Institute of Life and Health Engineering, Jinan University, Guangzhou 510632, China
| |
Collapse
|
4
|
Abstract
In preclinical studies, protein kinase C (PKC) enzymes have been implicated in regulating many aspects of pancreatic cancer development and progression. However, clinical Phase I or Phase II trials with compounds targeting classical PKC isoforms were not successful. Recent studies implicate that mainly atypical and novel PKC enzymes regulate oncogenic signaling pathways in pancreatic cancer. Members of these two subgroups converge signaling induced by mutant Kras, growth factors and inflammatory cytokines. Different approaches for the development of inhibitors for atypical PKC and novel PKC have been described; and new compounds include allosteric inhibitors and inhibitors that block ATP binding.
Collapse
Affiliation(s)
- Peter Storz
- Department of Cancer Biology, Mayo Clinic, Griffin Building, Room 306, 4500 San Pablo Road, Jacksonville, FL 32224, USA
| |
Collapse
|
5
|
Azijli K, Weyhenmeyer B, Peters GJ, de Jong S, Kruyt FAE. Non-canonical kinase signaling by the death ligand TRAIL in cancer cells: discord in the death receptor family. Cell Death Differ 2013; 20:858-868. [PMID: 23579241 PMCID: PMC3679459 DOI: 10.1038/cdd.2013.28] [Citation(s) in RCA: 144] [Impact Index Per Article: 12.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2013] [Revised: 03/06/2013] [Accepted: 03/07/2013] [Indexed: 12/14/2022] Open
Abstract
Tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-based therapy is currently evaluated in clinical studies as a tumor cell selective pro-apoptotic approach. However, besides activating canonical caspase-dependent apoptosis by binding to TRAIL-specific death receptors, the TRAIL ligand can activate non-canonical cell survival or proliferation pathways in resistant tumor cells through the same death receptors, which is counterproductive for therapy. Even more, recent studies indicate metastases-promoting activity of TRAIL. In this review, the remarkable dichotomy in TRAIL signaling is highlighted. An overview of the currently known mechanisms involved in non-canonical TRAIL signaling and the subsequent activation of various kinases is provided. These kinases include RIP1, IκB/ NF-κB, MAPK p38, JNK, ERK1/2, MAP3K TAK1, PKC, PI3K/Akt and Src. The functional consequences of their activation, often being stimulation of tumor cell survival and in some cases enhancement of their invasive behavior, are discussed. Interestingly, the non-canonical responses triggered by TRAIL in resistant tumor cells resemble that of TRAIL-induced signals in non-transformed cells. Better knowledge of the mechanism underlying the dichotomy in TRAIL receptor signaling may provide markers for selecting patients who will likely benefit from TRAIL-based therapy and could provide a rationalized basis for combination therapies with TRAIL death receptor-targeting drugs.
Collapse
Affiliation(s)
- K Azijli
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - B Weyhenmeyer
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - G J Peters
- Department of Medical Oncology, VU University Medical Center, Amsterdam, The Netherlands
| | - S de Jong
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| | - F A E Kruyt
- Department of Medical Oncology, University of Groningen, University Medical Center Groningen, Groningen, The Netherlands
| |
Collapse
|
6
|
PKC activator therapeutic for mild traumatic brain injury in mice. Neurobiol Dis 2010; 41:329-37. [PMID: 20951803 DOI: 10.1016/j.nbd.2010.10.001] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/14/2010] [Revised: 09/25/2010] [Accepted: 10/02/2010] [Indexed: 01/08/2023] Open
Abstract
Traumatic brain injury (TBI) is a frequent consequence of vehicle, sport and war related injuries. More than 90% of TBI patients suffer mild injury (mTBI). However, the pathologies underlying the disease are poorly understood and treatment modalities are limited. We report here that in mice, the potent PKC activator bryostatin1 protects against mTBI induced learning and memory deficits and reduction in pre-synaptic synaptophysin and post-synaptic spinophylin immunostaining. An effective treatment has to start within the first 8h after injury, and includes 5 × i.p. injections over a period of 14 days. The treatment is dose dependent. Exploring the effects of the repeated bryostatin1 treatment on the processing of the amyloid precursor protein, we found that the treatment induced an increase in the putative α-secretase ADAM10 and a reduction in β-secretase activities. Both these effects could contribute towards a reduction in β-amyloid production. These results suggest that bryostatin1 protects against mTBI cognitive and synaptic sequela by rescuing synapses, which is possibly mediated by an increase in ADAM10 and a decrease in BACE1 activity. Since bryostatin1 has already been extensively used in clinical trials as an anti-cancer drug, its potential as a remedy for the short- and long-term TBI sequelae is quite promising.
Collapse
|
7
|
Min Y, Shi J, Zhang Y, Liu S, Liu Y, Zheng D. Death receptor 5-recruited raft components contributes to the sensitivity of Jurkat leukemia cell lines to TRAIL-induced cell death. IUBMB Life 2009; 61:261-7. [PMID: 19242990 DOI: 10.1002/iub.166] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
Abstract
In the present study we demonstrated Jurkat leukemia cell lines of TIB152 and TIB153 with different sensitivities to recombinant soluble TRAIL cytotoxicity. TRAIL receptor death receptor 5 (DR5) was constitutively localized in the rafts in both cell lines. FADD, caspase-8, and PI3K-p85 subunit were recruited into DR5 lipid rafts of TIB152 but not in TIB153 cells. The expression and enzyme activity of acid sphingomyelinase, which digests sphingomyeline to produce ceramide and plays an essential role in lipid raft assembling, were higher in the rafts of TIB152 than in TIB153. These data provide evidences that DR5-recruited raft components contribute to the different sensitivity of Jurkat leukemia cell lines to TRAIL-induced cell death and may throw some light on the development of better therapeutic strategies for the cancer cells resistant to TRAIL treatment.
Collapse
Affiliation(s)
- Yifan Min
- National Laboratory of Medical Molecular Biology, Institute of Basic Medical Sciences, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing, China
| | | | | | | | | | | |
Collapse
|
8
|
Granulocyte colony-stimulating factor delays neutrophil apoptosis by inhibition of calpains upstream of caspase-3. Blood 2008; 112:2046-54. [PMID: 18524991 DOI: 10.1182/blood-2008-04-149575] [Citation(s) in RCA: 96] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023] Open
Abstract
Neutrophils have a very short life span and undergo apoptosis within 24 hours after leaving the bone marrow. Granulocyte colony-stimulating factor (G-CSF) is essential for the recruitment of fresh neutrophils from the bone marrow but also delays apoptosis of mature neutrophils. To determine the mechanism by which G-CSF inhibits neutrophil apoptosis, the kinetics of neutrophil apoptosis during 24 hours in the absence or presence of G-CSF were analyzed in vitro. G-CSF delayed neutrophil apoptosis for approximately 12 hours and inhibited caspase-9 and -3 activation, but had virtually no effect on caspase-8 and little effect on the release of proapoptotic proteins from the mitochondria. However, G-CSF strongly inhibited the activation of calcium-dependent cysteine proteases calpains, upstream of caspase-3, via apparent control of Ca(2+)-influx. Calpain inhibition resulted in the stabilization of the X-linked inhibitor of apoptosis (XIAP) and hence inhibited caspase-9 and -3 in human neutrophils. Thus, neutrophil apoptosis is controlled by G-CSF after initial activation of caspase-8 and mitochondrial permeabilization by the control of postmitochondrial calpain activity.
Collapse
|
9
|
Basu A, Sivaprasad U. Protein kinase Cepsilon makes the life and death decision. Cell Signal 2007; 19:1633-42. [PMID: 17537614 PMCID: PMC1986651 DOI: 10.1016/j.cellsig.2007.04.008] [Citation(s) in RCA: 117] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/30/2007] [Accepted: 04/23/2007] [Indexed: 12/20/2022]
Abstract
Cancer is caused by dysregulation in cellular signaling systems that control cell proliferation, differentiation and cell death. Protein kinase C (PKC), a family of serine/threonine kinases, plays an important role in the growth factor signal transduction pathway. PKCepsilon, however, is the only PKCepsilon isozyme that has been considered as an oncogene. It can contribute to malignancy by enhancing cell proliferation or by inhibiting cell death. This review focuses on how PKCepsilon collaborates with other signaling pathways, such as Ras/Raf/ERK and Akt, to regulate cell survival and cell death. We have also discussed how PKCepsilon mediates its antiapoptotic signal by altering the level or function of pro- and antiapoptotic Bcl-2 family members.
Collapse
Affiliation(s)
- Alakananda Basu
- Department of Molecular Biology and Immunology, University of North Texas Health Science Center, Fort Worth, TX 76107, USA.
| | | |
Collapse
|
10
|
Burkitt M, Magee C, O'Connor D, Campbell F, Cornford P, Greenhalf W. Potentiation of chemotherapeutics by the Hsp90 antagonist geldanamycin requires a steady serum condition. Mol Carcinog 2007; 46:466-75. [PMID: 17219417 DOI: 10.1002/mc.20296] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Inhibition of Hsp90 potentiates diverse chemotherapeutics, but it is not clear if this applies only to specific agents, tumor types or conditions. The aim of this report is to determine the effect of serum starvation (SS) on potentiation. SUIT2 cells were cultured with and without the presence of serum and 3-[4,5-dimethylthiazol-2-yl]-2,5-diphenyltetrazolium bromide (MTT) assays were carried out at time intervals. Cytotoxic agents were added individually or in combination. Immunohistochemistry of tumor samples and immunofluorescence of cultured cells were used to examine Hsp90 localization. In the presence of serum an at least additive effect of combining the Hsp90 inhibitor geldanamycin (GA) with 5-fluorouracil (5FU) was demonstrated. Following pretreatment with GA, 5FU and GA were synergistic. However, during SS GA was protective against 5FU. Geldanamycin also protected cells from 12-O-tetradecanoylphorbol-13-acetate (TPA) during SS. Protection of cells is transitory, as after 24 h of SS GA again has an at least additive negative effect on vitality with 5FU or TPA. Serum starvation of pancreatic cancer cell lines causes normally largely cytoplasmic Hsp90 to become predominantly nuclear localized. Hsp90 nuclear localization was observed in pancreatic and prostate tumors. Hsp90 binding to a pro-apoptotic client could explain the transitory protection of cells by Hsp90 inhibition during SS. Although potentiation of chemotherapeutics by Hsp90 inhibition is probably a general phenomenon, design of clinical trials should take into account that continuous co-administration may be ineffective because of a balance of synergy of the drugs in some cells and mutual inhibition of the two drug activities in other cells.
Collapse
Affiliation(s)
- Michael Burkitt
- Division of Gastroenterology, Liverpool University, Liverpool, United Kingdom
| | | | | | | | | | | |
Collapse
|
11
|
Gong J, Yang D, Kohanim S, Humphreys R, Broemeling L, Kurzrock R. Novel in vivo imaging shows up-regulation of death receptors by paclitaxel and correlates with enhanced antitumor effects of receptor agonist antibodies. Mol Cancer Ther 2006; 5:2991-3000. [PMID: 17148761 DOI: 10.1158/1535-7163.mct-06-0188] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
Susceptibility to apoptosis by tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) is mediated through cognate death receptor signaling. We hypothesized that auto-amplification of this apparatus would enhance antitumor effects in vivo and could be optimized using the results obtained from novel imaging techniques. We therefore imaged mice bearing human colorectal cancer (Colo205) tumor xenografts with HGS-ETR1 and HGS-ETR2 agonist antibodies to TRAIL receptor-1 (TRAIL-R1) and TRAIL-R2, respectively, after radiolabeling the antibodies. Paclitaxel significantly increased in vivo expression of TRAIL-R1 and TRAIL-R2 in a time-dependent manner. The imaging results were confirmed by immunoblots for steady-state protein levels (>20-fold increase in TRAIL-R1 and TRAIL-R2 levels in tumor xenografts by 48 h after paclitaxel administration). TRAIL-R1 and TRAIL-R2 mRNA expression did not change, suggesting that these effects were posttranscriptional. Sequential treatment with paclitaxel followed by HGS-ETR1 or HGS-ETR2 after 48 h resulted in markedly enhanced antitumor activity against Colo205 mouse xenografts. Our experiments suggest that sequential taxane treatment followed by TRAIL-R agonist antibodies could be applied in the clinic, and that novel imaging techniques using radiolabeled receptor antibodies may be exploitable to optimize sequence timing and patient selection.
Collapse
Affiliation(s)
- Jing Gong
- Department of Experimental Therapeutics, University of Texas M.D. Anderson Cancer Center, Box 4221515, Holcombe Boulevard, Houston, TX 77230-1402, USA
| | | | | | | | | | | |
Collapse
|
12
|
Dermitzaki E, Tsatsanis C, Charalampopoulos I, Androulidaki A, Alexaki VI, Castanas E, Gravanis A, Margioris AN. Corticotropin-releasing hormone activates protein kinase C in an isoenzyme-specific manner. Biochem Biophys Res Commun 2005; 327:828-36. [PMID: 15649420 DOI: 10.1016/j.bbrc.2004.12.078] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2004] [Indexed: 11/26/2022]
Abstract
Protein kinase C (PKC) has recently emerged as mediator of corticotropin-releasing hormone (CRH) effects. Aim of the present study was to study the effects of CRH on each PKC isoenzyme. As a model we have used the PC12 rat pheochromocytoma cell line, expressing the CRH type 1 receptor (CRHR1). Our data were as follows: (a) CRH-induced rapid phosphorylation of conventional PKCalpha and PKCbeta, accompanied by parallel increase of their concentration within nucleus. (b) CRH suppressed the phosphorylation of novel PKCdelta and PKCtheta;, which remained in the cytosol. (c) CRH-induced transient phosphorylation of atypical PKClambda and had no effect on PKCmu. (d) The effect of CRH on each PKC isoenzyme was blocked by a CRHR1 antagonist. (e) Blockade of conventional PKC phosphorylation inhibited CRH-induced calcium ion mobilization from intracellular stores as well as the CRH-induced apoptosis and Fas ligand production. In conclusion, our findings suggest that CRH via its CRHR1 receptor differentially regulates PKC-isoenzyme phosphorylation, an apparently physiologically relevant effect since blockade of conventional PKC phosphorylation abolished the biological effect of CRH.
Collapse
Affiliation(s)
- Erini Dermitzaki
- Department of Clinical Chemistry-Biochemistry, School of Medicine, University of Crete, Heraklion, Crete GR-710 03, Greece
| | | | | | | | | | | | | | | |
Collapse
|
13
|
Conesa-Zamora P, Mollinedo F, Corbalán-García S, Gómez-Fernández JC. A comparative study of the effect of the antineoplastic ether lipid 1-O-octadecyl-2-O-methyl-glycero-3-phosphocholine and some homologous compounds on PKCα and PKCɛ. Biochim Biophys Acta Mol Cell Biol Lipids 2005; 1687:110-9. [PMID: 15708359 DOI: 10.1016/j.bbalip.2004.11.008] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/19/2004] [Revised: 11/11/2004] [Accepted: 11/11/2004] [Indexed: 11/25/2022]
Abstract
The effects of the anti-neoplastic ether lipid ET-18-OCH3 and some structural homologues on the activity of protein kinase C alpha (PKC alpha) were studied and compared with the effects the same had on the activity of PKC epsilon. ET-18-OCH3 progressively inhibited the activity of PKC alpha as the concentration was increased up to 30 mol% of the total lipid, above which the effect was one of activation. The experiments carried out with the homologues showed that the methoxy group bound at the sn-2 position of the glycerol of ET-18-OCH3 is essential for both the initial inhibitory effect and the subsequent activation effect. On the other hand, variations in the type of bond linking substitutions in the sn-1 position, ether or ester, do not seem to play an important role in determining the activity of the enzyme. The effects were different on PKC epsilon since ET-18-OCH3 had a triphasic effect, activating the enzyme at low concentrations, inhibiting it at slightly higher concentrations and then activating it again at higher concentrations. In this case, when the homologues were used, it was observed that the presence of the methoxy group linked to the sn-2 position of glycerol and the type of bond linking substitutions to the sn-1 position were important for activating the enzyme, so that only homologues with ester bonds as LPC and PAPC were able to induce the initial activation step in a way similar to ET-18-OCH3. Substitution of the phosphocholine group of ET-18-OCH3 by phosphoserine led to a greater activation of PKC alpha, an effect that comes from the Ca(2+)-phospholipid binding site probably because of the specific interaction of this site with the phosphoserine group. The action of ET-18-OCH3 and its homologues, as demonstrated in this paper, may permit the selective inhibition or activation of PKC alpha and PKC epsilon by using the most suitable range of concentrations.
Collapse
Affiliation(s)
- Pablo Conesa-Zamora
- Departamento de Bioquímica y Biología Molecular A, Facultad de Veterinaria, Universidad de Murcia, Apartado de Correos 4021, E-30080-Murcia, Spain
| | | | | | | |
Collapse
|
14
|
Baryza JL, Brenner SE, Craske ML, Meyer T, Wender PA. Simplified Analogs of Bryostatin with Anticancer Activity Display Greater Potency for Translocation of PKCδ-GFP. ACTA ACUST UNITED AC 2004; 11:1261-7. [PMID: 15380186 DOI: 10.1016/j.chembiol.2004.06.014] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/20/2004] [Revised: 06/21/2004] [Accepted: 06/28/2004] [Indexed: 11/19/2022]
Abstract
Structurally simplified analogs of bryostatin 1, a marine natural product in clinical trials for the treatment of cancer, have been shown to be up to 50 times more potent than bryostatin 1 at inducing the translocation of PKCdelta-GFP from the cytosol of rat basophilic leukemia (RBL) cells. The end distribution of the protein is similar for all three compounds, despite a significant difference in translocation kinetics. The potency of the compounds for inducing the translocation response appears to be only qualitatively related to their binding affinity for PKC, highlighting the importance of using binding affinity in conjunction with real-time measurements of protein localization for the pharmacological profiling of biologically active agents.
Collapse
Affiliation(s)
- Jeremy L Baryza
- Department of Chemistry, Stanford University, Stanford, CA 94305, USA
| | | | | | | | | |
Collapse
|