1
|
Li J, Chang WT, Li CQ, Lee C, Huang HH, Hsu CW, Chen WJ, Zhu X, Wang CZ, Vanden Hoek TL, Shao ZH. Baicalein Preventive Treatment Confers Optimal Cardioprotection by PTEN/Akt/NO Activation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2017; 45:987-1001. [PMID: 28760044 DOI: 10.1142/s0192415x17500525] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Baicalein is a flavonoid with excellent oxidant scavenging capability. It has been reported to protect against a variety of oxidative injuries including ischemia/reperfusion (I/R). However, the optimal treatment strategy for I/R injury and the protective mechanisms are not fully understood. In this study we employed an established chick cardiomyocyte model of I/R and investigated the effects of three baicalein treatment strategies on reactive oxygen species (ROS) scavenging, nitric oxide (NO) production and cell viability. The molecular signaling pathways were also explored. Compared to the I/R control (cell death 52.2[Formula: see text][Formula: see text][Formula: see text]2.0%), baicalein preventive treatment (25[Formula: see text][Formula: see text]M, pretreated for 72[Formula: see text]h and continued through I/R) conferred the best protection (19.5[Formula: see text][Formula: see text][Formula: see text]3.9%, [Formula: see text]), followed by I/R treatment (treated during I/R) and reperfusion treatment (treated at reperfusion only). Preventive and I/R treatments almost completely abolished ROS generation during both ischemic and reperfusion phases, and increased NO production and Akt phosphorylation. Reperfusion treatment reduced the ROS burst in the early reperfusion phase only, and had no effect on NO production and Akt activation. Further, the phosphorylation of phosphatase and tensin homolog (PTEN), a phosphatase negatively regulating Akt activation, was significantly increased by baicalein preventive treatment and slightly by the I/R treatment. PTEN protein expression was reduced in the same trend accordingly. Baicalein reperfusion treatment had no effects on PTEN phosphorylation and expression. Our results indicate that baicalein preventive treatment confers optimal cardioprotection against I/R injury, and this protection involves effective oxidant scavenging and the activation of PTEN/Akt/NO pathway.
Collapse
Affiliation(s)
- Jing Li
- * Institute of Precision Medicine, Jining Medical University, Jining 272067, China.,† Department of Emergency Medicine, Center for Advanced Resuscitation Medicine and Program in Sudden Cardiac Death, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, University of Illinois at Chicago, IL 60612, USA
| | - Wei-Tien Chang
- † Department of Emergency Medicine, Center for Advanced Resuscitation Medicine and Program in Sudden Cardiac Death, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, University of Illinois at Chicago, IL 60612, USA.,‡ Department of Emergency Medicine, National Taiwan University College of Medicine and National Taiwan University Hospital, Taipei, Taiwan, R.O.C
| | - Chang-Qing Li
- † Department of Emergency Medicine, Center for Advanced Resuscitation Medicine and Program in Sudden Cardiac Death, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, University of Illinois at Chicago, IL 60612, USA
| | - Chunpei Lee
- † Department of Emergency Medicine, Center for Advanced Resuscitation Medicine and Program in Sudden Cardiac Death, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, University of Illinois at Chicago, IL 60612, USA
| | - Hsien-Hao Huang
- † Department of Emergency Medicine, Center for Advanced Resuscitation Medicine and Program in Sudden Cardiac Death, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, University of Illinois at Chicago, IL 60612, USA.,§ Department of Emergency Medicine, Taipei Veterans General Hospital and Emergency Medicine, College of Medicine, National Yang-Ming University, Taipei, Taiwan, R.O.C
| | - Chin-Wan Hsu
- † Department of Emergency Medicine, Center for Advanced Resuscitation Medicine and Program in Sudden Cardiac Death, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, University of Illinois at Chicago, IL 60612, USA.,¶ Department of Emergency Medicine, School of Medicine, College of Medicine; Department of Emergency and Critical Medicine, Wan-Fang Hospital, Taipei Medical University, Taipei, Taiwan, R.O.C
| | - Wen-Jone Chen
- ‡ Department of Emergency Medicine, National Taiwan University College of Medicine and National Taiwan University Hospital, Taipei, Taiwan, R.O.C
| | - Xiangdong Zhu
- ∥ Tang Center for Herbal Medicine Research and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA
| | - Chong-Zhi Wang
- ∥ Tang Center for Herbal Medicine Research and Department of Anesthesia & Critical Care, University of Chicago, Chicago, IL 60637, USA
| | - Terry L Vanden Hoek
- † Department of Emergency Medicine, Center for Advanced Resuscitation Medicine and Program in Sudden Cardiac Death, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, University of Illinois at Chicago, IL 60612, USA
| | - Zuo-Hui Shao
- † Department of Emergency Medicine, Center for Advanced Resuscitation Medicine and Program in Sudden Cardiac Death, Center for Cardiovascular Research, University of Illinois Hospital & Health Sciences System, University of Illinois at Chicago, IL 60612, USA
| |
Collapse
|
2
|
Huang HH, Shao ZH, Li CQ, Vanden Hoek TL, Li J. Baicalein Protects Cardiomyocytes Against Mitochondrial Oxidant Injury Associated with JNK Inhibition and Mitochondrial Akt Activation. THE AMERICAN JOURNAL OF CHINESE MEDICINE 2014; 42:79-94. [DOI: 10.1142/s0192415x14500050] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
Baicalein, a flavonoid derived from Scutellaria baicalensis Georgi, possesses cardioprotection against oxidant injury by scavenging reactive oxygen species (ROS). Few studies investigate whether baicalein protection is mediated by attenuating mitochondrial ROS and modulating the prosurvival and proapoptotic signaling. Primary cultured chick cardiomyocytes were used to study the role of baicalein in mitochondrial superoxide [Formula: see text] generation and signaling of Akt and JNK. Cells were exposed to H 2 O 2 for 2 h and baicalein was given 2 h prior to and during 2 h of H 2 O 2 exposure. Cell viability was assessed by propidium iodide and DNA fragmentation. H 2 O 2 (500 μM) significantly induced 45.3 ± 6.2% of cell death compared to the control (p < 0.001) and resulted in DNA laddering. Baicalein (10, 25 or 50 μM) dose-dependently reduced the cell death to 38.7 ± 5.6% (p = 0.226); 31.2 ± 3.9% (p < 0.01); 30.3 ± 5.3% (p < 0.01), respectively. It also attenuated DNA laddering. Further, baicalein decreased intracellular ROS and mitochondrial [Formula: see text] generation that was confirmed by superoxide dismutase PEG-SOD and mitochondria electron transport chain complex III inhibitor stigmatellin. In addition, baicalein increased Akt phosphorylation and decreased JNK phosphorylation in H 2 O 2-exposed cells. Moreover, baicalein augmented mitochondrial phosphorylation of Akt Thr308 and GSK3β Ser9, and prevented mitochondrial cytochrome c release assessed by cellular fractionation. Our results suggest that baicalein cardioprotection may involve an attenuation of mitochondrial [Formula: see text] and an increase in mitochondrial phosphorylation of Akt and GSK3β while decreasing JNK activation.
Collapse
Affiliation(s)
- Hsien-Hao Huang
- Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital and Health Sciences System, Chicago, IL 60612, USA
- Department of Emergency Medicine, Taipei Veterans General Hospital and Emergency Medicine, College of Medicine, National Yang-Ming University, Taipei, Taiwan
| | - Zuo-Hui Shao
- Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital and Health Sciences System, Chicago, IL 60612, USA
| | - Chang-Qing Li
- Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital and Health Sciences System, Chicago, IL 60612, USA
| | - Terry L. Vanden Hoek
- Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital and Health Sciences System, Chicago, IL 60612, USA
| | - Jing Li
- Department of Emergency Medicine, Center for Cardiovascular Research, University of Illinois Hospital and Health Sciences System, Chicago, IL 60612, USA
| |
Collapse
|
3
|
Yongxiang W, Liang G, Qinshu S. Apoptosis of human pancreatic carcinoma PC-2 cells by an antisense oligonucleotide specific to point mutated K-ras. Pathol Oncol Res 2014; 20:81-5. [PMID: 23828694 PMCID: PMC3882557 DOI: 10.1007/s12253-013-9661-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 12/19/2012] [Accepted: 06/03/2013] [Indexed: 12/15/2022]
Abstract
The prognosis of pancreatic carcinoma is poor due to the difficulty in early diagnosis, insensitivity to routine therapies and limited understanding of its pathological mechanisms. Gene therapy is now becoming an important strategy for the treatment of pancreatic carcinoma, which includes antisense gene therapy. In this study, we investigated the effect of an antisense oligonucleotide specific to point mutated K-ras on the apoptosis of human pancreatic carcinoma cells in vitro. Human pancreatic carcinoma PC-2 cells were transfected with an antisense oligonucleotide specific to a K-ras point mutation by liposomes. The effect of the antisense oligonucleotide on the apoptosis of PC-2 cells was studied using flow cytometry, TUNEL, and phase contrast microscopy. An apoptotic peak was observed in the experimental group, and most cells were arrested at the G1 phase with few cells at the S phase. The numbers of apoptotic cells in the experimental group increased as indicated by TUNEL and phase contrast microscopy. An antisense oligonucleotide specific to a K-ras point mutation promotes apoptosis in PC-2 cells in vitro perhaps by inhibition of ras gene expression.
Collapse
Affiliation(s)
- Wang Yongxiang
- Department of General Surgery, Zhejiang Provincial People’s Hospital, Hangzhou, 310014 China
| | - Gao Liang
- Department of Oncology, Zhejiang Provincial People’s Hospital, Hangzhou, 310014 China
| | - Shao Qinshu
- Department of General Surgery, Zhejiang Provincial People’s Hospital, Hangzhou, 310014 China
| |
Collapse
|
4
|
Marchand B, Tremblay I, Cagnol S, Boucher MJ. Inhibition of glycogen synthase kinase-3 activity triggers an apoptotic response in pancreatic cancer cells through JNK-dependent mechanisms. Carcinogenesis 2011; 33:529-37. [PMID: 22201186 DOI: 10.1093/carcin/bgr309] [Citation(s) in RCA: 52] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Recent evidences suggest that the activity of glycogen synthase kinase-3 (GSK3) contributes to the tumorigenic potential of pancreatic cancer cells through modulation of cell proliferation and survival. However, further investigations are needed to identify GSK3-dependent mechanisms involved in the control of pancreatic cancer cell proliferation and survival. This study was undertaken to provide further support for a role of GSK3 in pancreatic cancer cell growth as well as to identify new cellular and molecular mechanisms involved. Herein, we demonstrate that prolonged inhibition of GSK3 triggers an apoptotic response only in human pancreatic cancer cells but not in human non-transformed pancreatic epithelial cells. We show that prolonged inhibition of GSK3 activity increases Bim messenger RNA and protein expressions. Moreover, we provide evidence that activation of the c-jun N-terminal kinase (JNK) pathway is necessary for the GSK3 inhibition-mediated increase in Bim expression and apoptotic response. Finally, we demonstrate that concomitant inhibition of GSK3 potentiates the death ligand-induced apoptotic response in pancreatic cancer cells but not in non-transformed pancreatic epithelial cells and that this effect also requires JNK activity. Considering that different approaches leading to stimulation of death receptor signaling are under clinical trials for treatment of unresectable or metastatic pancreatic cancer, inhibition of GSK3 could represent an attractive new avenue to improve their effectiveness.
Collapse
Affiliation(s)
- Benoît Marchand
- Service de Gastroentérologie, Département de Médecine, Université de Sherbrooke, 3001, 12e avenue nord, Sherbrooke, Québec, Canada
| | | | | | | |
Collapse
|
5
|
Kwak CS, Yeo EJ, Moon SC, Kim YW, Ahn HJ, Park SC. Perilla Leaf, Perilla frutescens, Induces Apoptosis and G1 Phase Arrest in Human Leukemia HL-60 Cells Through the Combinations of Death Receptor-Mediated, Mitochondrial, and Endoplasmic Reticulum Stress-Induced Pathways. J Med Food 2009; 12:508-17. [DOI: 10.1089/jmf.2008.1103] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Affiliation(s)
- Chung Shil Kwak
- Institute on Aging, Medical School, Seoul National University, Seoul
| | - Eui Ju Yeo
- Department of Biochemistry, Gachon Medical School, Incheon, Republic of Korea
| | - Sung Chae Moon
- Institute on Aging, Medical School, Seoul National University, Seoul
| | - Young Wha Kim
- Institute on Aging, Medical School, Seoul National University, Seoul
| | - Hong Ju Ahn
- The Aging and Apoptosis Research Center, Medical School, Seoul National University, Seoul
| | - Sang Chul Park
- The Aging and Apoptosis Research Center, Medical School, Seoul National University, Seoul
| |
Collapse
|
6
|
d-δ-Tocotrienol-mediated suppression of the proliferation of human PANC-1, MIA PaCa-2, and BxPC-3 pancreatic carcinoma cells. Pancreas 2009; 38:e124-36. [PMID: 19346993 DOI: 10.1097/mpa.0b013e3181a20f9c] [Citation(s) in RCA: 68] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
OBJECTIVE The rate-limiting activity of the mevalonate pathway, 3-hydroxy-3-methylglutaryl coenzyme A (HMG CoA) reductase, provides intermediates essential for growth. Competitive inhibitors of HMG CoA reductase, such as the statins, and down-regulators of reductase, such as the tocotrienols, suppress tumor growth. We evaluated the impact of d-delta-tocotrienol, the most potent vitamin E isomer, on human MIA PaCa-2 and PANC-1 pancreatic carcinoma cells and BxPC-3 pancreatic ductal adenocarcinoma cells. METHODS Cell proliferation was measured by using CellTiter 96 Aqueous One Solution (Promega, Madison, Wis). Cell cycle distribution was determined by flow cytometry. Apoptosis was evaluated by Annexin V staining and fluorescence microscopy after dual staining with acridine orange and ethidium bromide. RESULTS d-delta-Tocotrienol induced concentration-dependent suppression of cell proliferation with 50% inhibitory concentrations of 28 (6) micromol/L (MIA PaCa-2), 35 (7) micromol/L (PANC-1), and 35 (8) microL (BxPC-3), respectively. These effects are attributable to cell cycle arrest at the G1 phase and apoptosis. Mevalonate attenuated d-delta-tocotrienol-mediated growth inhibition. A physiologically attainable blend of d-delta-tocotrienol and lovastatin synergistically suppressed the proliferation of MIA PaCa-2 cells. CONCLUSIONS Suppression of mevalonate pathway activities, be it by modulators of HMG CoA reductase (statins, tocotrienols, and farnesol), farnesyl transferase (farnesyl transferase inhibitors), and/or mevalonate pyrophosphate decarboxylase (phenylacetate) activity, may have a potential in pancreatic cancer chemotherapy.
Collapse
|
7
|
Inhibition of c-Jun N-terminal kinase sensitizes tumor cells to flavonoid-induced apoptosis through down-regulation of JunD. Toxicol Appl Pharmacol 2008; 227:468-76. [DOI: 10.1016/j.taap.2007.11.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2007] [Revised: 10/30/2007] [Accepted: 11/07/2007] [Indexed: 11/21/2022]
|
8
|
Kim HS, Kim JW, Gang J, Wen J, Koh SS, Koh JS, Chung HH, Song SY. The farnesyltransferase inhibitor, LB42708, inhibits growth and induces apoptosis irreversibly in H-ras and K-ras-transformed rat intestinal epithelial cells. Toxicol Appl Pharmacol 2006; 215:317-29. [PMID: 16712893 DOI: 10.1016/j.taap.2006.03.011] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2005] [Revised: 03/20/2006] [Accepted: 03/22/2006] [Indexed: 02/04/2023]
Abstract
LB42708 (LB7) and LB42908 (LB9) are pyrrole-based orally active farnesyltransferase inhibitors (FTIs) that have similar structures. The in vitro potencies of these compounds against FTase and GGTase I are remarkably similar, and yet they display different activity in apoptosis induction and morphological reversion of ras-transformed rat intestinal epithelial (RIE) cells. Both FTIs induced cell death despite K-ras prenylation, implying the participation of Ras-independent mechanism(s). Growth inhibition by these two FTIs was accompanied by G1 and G2/M cell cycle arrests in H-ras and K-ras-transformed RIE cells, respectively. We identified three key markers, p21(CIP1/WAF1), RhoB and EGFR, that can explain the differences in the molecular mechanism of action between two FTIs. Only LB7 induced the upregulation of p21(CIP1/WAF1) and RhoB above the basal level that led to the cell cycle arrest and to distinct morphological alterations of ras-transformed RIE cells. Both FTIs successfully inhibited the ERK and activated JNK in RIE/K-ras cells. While the addition of conditioned medium from RIE/K-ras reversed the growth inhibition of ras-transformed RIE cells by LB9, it failed to overcome the growth inhibitory effect of LB7 in both H-ras- and K-ras-transformed RIE cells. We found that LB7, but not LB9, decreased the expression of EGFRs that confers the cellular unresponsiveness to EGFR ligands. These results suggest that LB7 causes the induction of p21(CIP1/WAF1) and RhoB and downregulation of EGFR that may serve as critical steps in the mechanism by which FTIs trigger irreversible inhibitions on the cell growth and apoptosis in ras-transformed cells.
Collapse
Affiliation(s)
- Han-Soo Kim
- Brain Korea 21 Project for Medical Science, Yonsei University College of Medicine, Seoul, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
9
|
Pei XY, Dai Y, Rahmani M, Li W, Dent P, Grant S. The farnesyltransferase inhibitor L744832 potentiates UCN-01-induced apoptosis in human multiple myeloma cells. Clin Cancer Res 2005; 11:4589-600. [PMID: 15958645 DOI: 10.1158/1078-0432.ccr-04-2346] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE The purpose of this study was to characterize interactions between the farnesyltransferase inhibitor L744832 and the checkpoint abrogator UCN-01 in drug-sensitive and drug-resistant human myeloma cell lines and primary CD138+ multiple myeloma cells. EXPERIMENTAL DESIGN Wild-type and drug-resistant myeloma cell lines were exposed to UCN-01 +/- L744832 for 24 hours, after which mitochondrial injury, caspase activation, apoptosis, and various perturbations in signaling and survival pathways were monitored. RESULTS Simultaneous exposure of myeloma cells to marginally toxic concentrations of L744832 and UCN-01 resulted in a synergistic induction of mitochondrial damage, caspase activation, and apoptosis, associated with activation of p34cdc2 and c-Jun-NH2-kinase and inactivation of extracellular signal-regulated kinase, Akt, GSK-3, p70(S6K), and signal transducers and activators of transcription 3 (STAT3). Enhanced lethality for the combination was also observed in primary CD138+ myeloma cells, but not in their CD138- counterparts. L744832/UCN-01-mediated lethality was not attenuated by conventional resistance mechanisms to cytotoxic drugs (e.g., melphalan or dexamethasone), addition of exogenous interleukin-6 or insulin-like growth factor-I, or the presence of stromal cells. In contrast, enforced activation of STAT3 significantly protected myeloma cells from L744832/UCN-01-induced apoptosis. CONCLUSIONS Coadministration of the farnesyltransferase inhibitor L744832 promotes UCN-01-induced apoptosis in human multiple myeloma cells through a process that may involve perturbations in various survival signaling pathways, including extracellular signal-regulated kinase, Akt, and STAT3, and through a process capable of circumventing conventional modes of myeloma cell resistance, including growth factor- and stromal cell-related mechanisms. They also raise the possibility that combined treatment with farnesyltransferase inhibitors and UCN-01 could represent a novel therapeutic strategy in multiple myeloma.
Collapse
Affiliation(s)
- Xin-Yan Pei
- Department of Medicine, Virginia Commonwealth University/Medical College of Virginia, Richmond, Virginia 23298, USA
| | | | | | | | | | | |
Collapse
|
10
|
Tabernero J, Rojo F, Marimón I, Voi M, Albanell J, Guix M, Vázquez F, Carulla J, Cooper M, Andreu J, Van Vreckem A, Bellmunt J, Manne V, Manning JA, Garrido C, Felip E, Del Campo JM, García M, Valverde S, Baselga J. Phase I Pharmacokinetic and Pharmacodynamic Study of Weekly 1-Hour and 24-Hour Infusion BMS-214662, a Farnesyltransferase Inhibitor, in Patients With Advanced Solid Tumors. J Clin Oncol 2005; 23:2521-33. [PMID: 15710949 DOI: 10.1200/jco.2005.00.398] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Purpose BMS-214662 is a potent, nonpeptide, small molecule inhibitor of human farnesyltransferase (FT). We have conducted a phase I pharmacokinetic (PK) and pharmacodynamic study of BMS-214662 administered intravenously weekly with 1- and 24-hour infusions. The objectives were to determine the dose-limiting toxicities and the recommended dose (RD), to describe PKs, and to evaluate the relationships between BMS-214662 exposure, FT inhibition, downstream signaling, and induction of apoptosis in tumor samples. Patients and Methods Patients with advanced solid tumors and adequate organ function were eligible. The dose was escalated according to a modified Fibonacci schedule. Results BMS-214662 was escalated from 56 to 278 mg/m2 in 37 patients in the 1-hour schedule, and from 84 to 492 mg/m2 in 31 patients in the 24-hour schedule. Dose-limiting toxicities included gastrointestinal and renal events. The RDs were 209 mg/m2 and 275 mg/m2 in the 1- and 24-hour schedules, respectively. Five patients (three with breast, one with gastric, and one with renal cell cancer) had clinical benefit from treatment. BMS-214662 exhibited linear PKs with area under the concentration-time curves at the RDs of 27 and 32 μM × h in the 1- and 24-hour schedules, respectively. The pattern of FT inhibition in peripheral-blood mononuclear cells at the RDs was different in the two schedules: high (> 80%) but short-lived (≤ 6 hours) in the 1-hour infusion and moderate (> 40%) but long-lived (24 hours) in the 24-hour infusion. BMS-214662 induced apoptosis in tumors but did not inhibit MAPK signaling. Conclusion BMS-214662 can be safely delivered in both the 1-hour and 24-hour infusions at biologically active doses, with the preclinical, PK, and pharmacodynamic profiles favoring the 24-hour schedule.
Collapse
Affiliation(s)
- Josep Tabernero
- Medical Oncology Department, Vall d'Hebron University Hospital, Universitat Autònoma de Barcelona, Barcelona, Spain. [corrected]
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
11
|
Lee HJ, Wang CJ, Kuo HC, Chou FP, Jean LF, Tseng TH. Induction apoptosis of luteolin in human hepatoma HepG2 cells involving mitochondria translocation of Bax/Bak and activation of JNK. Toxicol Appl Pharmacol 2005; 203:124-31. [PMID: 15710173 DOI: 10.1016/j.taap.2004.08.004] [Citation(s) in RCA: 124] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2004] [Accepted: 08/10/2004] [Indexed: 11/28/2022]
Abstract
Since hepatocellular carcinoma remains a major challenging clinical problem in many parts of the world including Eastern Asia and Southern Africa, it is imperative to develop more effective chemopreventive and chemotherapy agents. Herein, we present an investigation regarding the anticancer potential of luteolin, a natural flavonoid, and the mechanism of its action in human hepatoma HepG2 cells. Using DNA fragmentation assay and nuclear staining assay, it showed that luteolin induced apoptosis of HepG2 cells. Luteolin induced the cytosolic release of cytochrome c and activated CPP32. We found that Bax and Bak translocated to mitochondria apparently, whereas Fas ligand (FasL) was unchanged after a treatment with luteolin for 3 h. In addition, it showed that c-Jun NH2-terminal kinase (JNK) was activated after the treatment of luteolin for 3-12 h. Further investigation showed that a specific JNK inhibitor, SP600125, reduced the activation of CPP 32, the mitochondrial translocation of Bax, as well as the cytosolic release of cytochrome c that induced by luteolin. Finally, the apoptosis induced by luteolin was suppressed by a pretreatment with SP600125 via evaluating annexin V-FITC binding assay. These data suggest that luteolin induced apoptosis via mechanisms involving mitochondria translocation of Bax/Bak and activation of JNK.
Collapse
Affiliation(s)
- Herng-Jiun Lee
- Institute of Biochemistry, Chung Shan Medical University, Taichung, Taiwan
| | | | | | | | | | | |
Collapse
|
12
|
Mo H, Elson CE. Studies of the isoprenoid-mediated inhibition of mevalonate synthesis applied to cancer chemotherapy and chemoprevention. Exp Biol Med (Maywood) 2004; 229:567-85. [PMID: 15229351 DOI: 10.1177/153537020422900701] [Citation(s) in RCA: 233] [Impact Index Per Article: 11.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Pools of farnesyl diphosphate and other phosphorylated products of the mevalonate pathway are essential to the post-translational processing and physiological function of small G proteins, nuclear lamins, and growth factor receptors. Inhibitors of enzyme activities providing those pools, namely, 3-hydroxy-3-methylglutaryl coenzyme A (HMG CoA) reductase and mevalonic acid-pyrophosphate decarboxylase, and of activities requiring substrates from the pools, the prenyl protein transferases, have potential for development as novel chemotherapeutic agents. Their potentials as suggested by the clinical responses recorded in Phase I and II investigations of inhibitors of HMG CoA reductase (the statins), of mevalonic acid-pyrophosphate decarboxylase (sodium phenylacetate and sodium phenylbutyrate), and of farnesyl protein transferase (R115777, SCH66336, BMS-214662, Tipifarnib, L-778,123, and, prematurely, perillyl alcohol) are dimmed by dose-limiting toxicities. These nondiscriminant growth-suppressive agents induce G1 arrest and initiate apoptosis and differentiation, effects attributed to modulation of cell signaling pathways either by modulating gene expression, suppressing the post-translational processing of signaling proteins and growth factor receptors, or altering diacylglycerol signaling. Diverse isoprenoids and the HMG CoA reductase inhibitor, lovastatin, modulate cell growth, induce cell cycle arrest, initiate apoptosis, and suppress cellular signaling activities. Perillyl alcohol, the isoprenoid of greatest clinical interest, initially was considered to inhibit farnesyl protein transferase; follow-up studies revealed that perillyl alcohol suppresses the synthesis of small G proteins and HMG CoA reductase. In sterologenic tissues, sterol feedback control, mediated by sterol regulatory element binding proteins (SREBPs) 1a and 2, exerts the primary regulation on HMG CoA reductase activity at the transcriptional level. Secondary regulation, a nonsterol isoprenoid-mediated fine-tuning of reductase activity, occurs at the levels of reductase translation and degradation. HMG CoA reductase activity in tumors is elevated and resistant to sterol feedback regulation, possibly as a consequence of aberrant SREBP activities. Nonetheless, tumor reductase remains sensitive to isoprenoid-mediated post-transcriptional downregulation. Farnesol, an acyclic sesquiterpene, and farnesyl homologs, gamma-tocotrienol and various farnesyl derivatives, inhibit reductase synthesis and accelerate reductase degradation. Cyclic monoterpenes, d-limonene, menthol and perillyl alcohol and beta-ionone, a carotenoid fragment, lower reductase mass; perillyl alcohol and d-limonene lower reductase mass by modulating translational efficiency. The elevated reductase expression and greater demand for nonsterol products to maintain growth amplify the susceptibility of tumor reductase to isoprenoids, therein rendering tumor cells more responsive than normal cells to isoprenoid-mediated growth suppression. Blends of lovastatin, a potent nondiscriminant inhibitor of HMG CoA reductase, and gamma-tocotrienol, a potent isoprenoid shown to post-transcription-ally attenuate reductase activity with specificity for tumors, synergistically affect the growth of human DU145 and LNCaP prostate carcinoma cells and pending extensive preclinical evaluation, potentially offer a novel chemotherapeutic strategy free of the dose-limiting toxicity associated with high-dose lovastatin and other nondiscriminant mevalonate pathway inhibitors.
Collapse
Affiliation(s)
- Huanbiao Mo
- Department of Nutrition and Food Sciences, Texas Woman's University, Denton, TX 76204, USA.
| | | |
Collapse
|
13
|
Manne V, Lee FYF, Bol DK, Gullo-Brown J, Fairchild CR, Lombardo LJ, Smykla RA, Vite GD, Wen MLD, Yu C, Wong TW, Hunt JT. Apoptotic and Cytostatic Farnesyltransferase Inhibitors Have Distinct Pharmacology and Efficacy Profiles in Tumor Models. Cancer Res 2004; 64:3974-80. [PMID: 15173010 DOI: 10.1158/0008-5472.can-03-3849] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
BMS-214662 and BMS-225975 are tetrahydrobenzodiazepine-based farnesyltransferase inhibitors (FTIs) that have nearly identical structures and very similar pharmacological profiles associated with farnesyltransferase (FT) inhibition. Despite their similar activity against FT in vitro and in cells, these compounds differ dramatically in their apoptotic potency and tumor-regressing activity in vivo. BMS-214662 is the most potent apoptotic FTI known and exhibits curative responses in mice bearing a variety of staged human tumor xenografts such as HCT-116 human colon tumor. By contrast, BMS-225975 does not cause tumor regression and at best causes partial tumor growth inhibition in staged HCT-116 human colon tumor xenografts. Lack of tumor regression activity in BMS-225975 was attributable to its relatively weak apoptotic potency, not to poor cell permeability or pharmacokinetics. Both compounds were equally effective in inhibiting Ras processing and causing accumulation of a variety of nonfarnesylated substrates of FT in HCT-116 cells. Because BMS-225975 has poor apoptotic activity compared with BMS-214662 but inhibits FT to the same extent as BMS-214662, it is very unlikely that FT inhibition alone can account for the apoptotic potency of BMS-214662. Clearly distinct patterns of sensitivities in a cell line panel were obtained for the apoptotic FTI BMS-214662 and the cytostatic FTI BMS-225975. Activation of the c-Jun-NH(2)-terminal kinase pathway was readily observed with BMS-214662 but not with BMS-225975. We developed a highly sensitive San-1 murine xenograft tumor model that is particularly useful for evaluating the in vivo activity of cytostatic FTIs such as BMS-225975.
Collapse
Affiliation(s)
- Veeraswamy Manne
- Oncology Drug Discovery and Discovery Chemistry, Bristol-Myers Squibb Company Pharmaceutical Research Institute, Princeton, New Jersey 08543, USA.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Jones TH, Clark DA, Heterick BE, Snelling RR. Farnesylamine from the ant Monomorium fieldi Forel. JOURNAL OF NATURAL PRODUCTS 2003; 66:325-326. [PMID: 12662086 DOI: 10.1021/np020558f] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/24/2023]
Abstract
(2E)- and (2Z)-Farnesylamine were detected in the extracts of the myrmicine ant Monomorium fieldi Forel from Australia. Their structures were established by direct comparison with synthetic (2E)- and (2Z)-farnesylamine. This finding of a sesquiterpene is unique in a genus known to produce unbranched fatty acid derived alkaloids and amines. Additionally, while farnesylamine has not been reported from natural sources, the synthetic material has been shown to have a variety of biological activities.
Collapse
Affiliation(s)
- T H Jones
- Department of Chemistry, Virginia Military Institute, Lexington, Virginia 24450, USA.
| | | | | | | |
Collapse
|
15
|
Vervenne WL, Bos CL, Rens LS, Peppelenbosch MP, Richel DJ. Farnesyl Protein Transferase Inhibition Interferes with Activation of MAP Kinase Family Members in Human Peripheral Blood Monocytes. Mol Med 2002. [DOI: 10.1007/bf03402091] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/28/2022] Open
|