1
|
Xie Q, Zhang H, Wang M, Yan K, Hu F, Xu M. A model about regulation on three division modes of stem cell. J Theor Biol 2024; 581:111746. [PMID: 38280545 DOI: 10.1016/j.jtbi.2024.111746] [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: 10/11/2023] [Revised: 01/03/2024] [Accepted: 01/20/2024] [Indexed: 01/29/2024]
Abstract
We construct a multi-stage cell lineage model for cell division, apoptosis and movement. Cells are assumed to secrete and respond to negative feedback molecules which act as a control on the stem cell divisions (including self-renewal, asymmetrical cell division (ACD) and differentiation). The densities of cells and molecules are described by coupled reaction-diffusion partial differential equations, and the plane wavefront propagation speeds can be obtained analytically and verified numerically. It is found that with ACD the population and propagation of stem cells can be promoted but the negative regulation on self-renewal and differentiation will work slowly. Regulatory inhibition on differentiation will inversely increase stem cells but not affect the population and wave propagation of the cell lineage. While negative regulation on self-renewal and ACD will decrease the population of stem cells and slow down the propagation, and even drive stem cells to extinction. Moreover we find that inhibition on self-renewal has a strength advantage while inhibition on ACD has a range advantage to kill stem cells. Possible relations to model cancer development and therapy are also discussed.
Collapse
Affiliation(s)
- Qingxin Xie
- School of Science, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Han Zhang
- School of Science, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Maoxiang Wang
- School of Science, Nanjing University of Science and Technology, Nanjing 210094, China.
| | - Kexun Yan
- School of Science, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Fenglan Hu
- School of Science, Nanjing University of Science and Technology, Nanjing 210094, China
| | - Meng Xu
- School of Science, Nanjing University of Science and Technology, Nanjing 210094, China
| |
Collapse
|
2
|
Bazinet A, Kantarjian HM. Moving toward individualized target-based therapies in acute myeloid leukemia. Ann Oncol 2023; 34:141-151. [PMID: 36423744 DOI: 10.1016/j.annonc.2022.11.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/29/2022] [Revised: 10/24/2022] [Accepted: 11/09/2022] [Indexed: 11/22/2022] Open
Abstract
Acute myeloid leukemia (AML) is a heterogeneous disease at the genetic level. The field of AML therapy is increasingly shifting away from uniform approaches based solely on intensive chemotherapy (such as '7 + 3') toward personalized therapy. The treatment of AML can now be individualized based on patient characteristics and cytogenetic/molecular disease features. In this review, we provide a comprehensive updated summary of personalized, target-directed therapy in AML. We first discuss the selection of intensive versus low-intensity treatment approaches based on the patient's age and/or comorbidities. We follow with a detailed review of specific molecularly defined AML subtypes that benefit from the addition of targeted agents. In this context, we highlight the urgent need for novel therapies in tumor protein p53 (TP53)-mutated AML. We then propose approaches to optimize AML therapy in patients without directly actionable mutations. We conclude with a discussion on the emerging role of using measurable residual disease to modify therapy based on the quality of response.
Collapse
Affiliation(s)
- A Bazinet
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, USA
| | - H M Kantarjian
- Department of Leukemia, The University of Texas MD Anderson Cancer Center, Houston, USA.
| |
Collapse
|
3
|
Regulatory Action of all trans Retinoic Acid on Metastasis Induced lung Cell Metabolic Changes during Implantation of B16F10 Cancer Cells in C57BL6 Mice. JOURNAL OF PURE AND APPLIED MICROBIOLOGY 2021. [DOI: 10.22207/jpam.15.2.26] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
The changes that occur during metastasis lodging is under intense research now to develop preventive new drugs to fight against the deadly metastasis. The molecular drug, all trans Retinoic Acid (ATRA) has regulatory effects on signal mediated metabolism. In this study, we have analyzed the metastasis facilitating metabolic changes in mice lung when a highly metastatic melanoma cell line (B16F10) having potency to lodge in lung was implanted via tail vein injection into C57BL/6 mice (1×106 cells/ml in PBS). One group of implanted mice were treated with 0.60 mg of ATRA per Kg body weight daily for 21 days. The alteration of protein, enzymatic and non-enzymatic antioxidants (SOD, Catalase, GPX, GSH) levels and the lipid profile with cholesterol level were evaluated in the lung tissues. The ATRA treatment caused 62.16% inhibition on metastatic nodule formation. Compared to normal mice, the cancer control mice showed an increased (p≤ 0.01**) total protein, LPO and NO and a decreased antioxidant. In ATRA treated group, all these levels were reverted to near normal levels with a high significance (p≤ 0.01**) difference from untreated cancer mice. The lipid profile and cholesterol level also were altered in cancer and were normalized in ATRA treated group with high significance (p≤ 0.01**). All these results implies that the metabolic changes induced in the lung tissue during metastatic lodging of melanoma cells were prevented and regularized by the ATRA treatment in vivo which give a scope of anti-metastatic therapy using ATRA.
Collapse
|
4
|
Chowdhury T, Roymahapatra G, Mandal SM. In Silico Identification of a Potent Arsenic Based Approved Drug Darinaparsin against SARS-CoV-2: Inhibitor of RNA Dependent RNA polymerase (RdRp) and Essential Proteases. Infect Disord Drug Targets 2021; 21:608-618. [PMID: 32718300 DOI: 10.2174/1871526520666200727153643] [Citation(s) in RCA: 14] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2020] [Revised: 06/14/2020] [Accepted: 06/16/2020] [Indexed: 11/22/2022]
Abstract
BACKGROUND COVID-19 is a life-threatening novel corona viral infection to our civilization and spreading rapidly. Tremendousefforts have been made by the researchers to search for a drug to control SARS-CoV-2. METHODS Here, a series of arsenical derivatives were optimized and analyzed with in silico study to search the inhibitor of RNA dependent RNA polymerase (RdRp), the major replication factor of SARS-CoV-2. All the optimized derivatives were blindly docked with RdRp of SARS-CoV-2 using iGEMDOCK v2.1. RESULTS Based on the lower idock score in the catalytic pocket of RdRp, darinaparsin (-82.52 kcal/- mol) was revealed to be the most effective among them. Darinaparsin strongly binds with both Nsp9 replicase protein (-8.77 kcal/mol) and Nsp15 endoribonuclease (-8.3 kcal/mol) of SARS-- CoV-2 as confirmed from the AutoDock analysis. During infection, the ssRNA of SARS-CoV-2 is translated into large polyproteins forming viral replication complex by specific proteases like 3CL protease and papain protease. This is also another target to control the virus infection where darinaparsin also performs the inhibitory role to proteases of 3CL protease (-7.69 kcal/mol) and papain protease (-8.43 kcal/mol). CONCLUSION In the host cell, the furin protease serves as a gateway to the viral entry and darinaparsin docked with furin protease, which revealed a strong binding affinity. Thus, screening of potential arsenic drugs would help in providing the fast in-vitro to in-vivo analysis towards the development of therapeutics against SARS-CoV-2.
Collapse
Affiliation(s)
- Trinath Chowdhury
- Central Research Facility, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| | | | - Santi M Mandal
- Central Research Facility, Indian Institute of Technology Kharagpur, Kharagpur 721302, India
| |
Collapse
|
5
|
Zhang X, Liu L, Yao Y, Gong S, Wang M, Xi J, Chen L, Wei S, Zhang H, Zhao C, Wang H. Treatment of non-high-risk acute promyelocytic leukemia with realgar-indigo naturalis formula (RIF) and all-trans retinoid acid (ATRA): study protocol for a randomized controlled trial. Trials 2020; 21:7. [PMID: 31898521 PMCID: PMC6941314 DOI: 10.1186/s13063-019-3983-2] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2019] [Accepted: 12/10/2019] [Indexed: 11/10/2022] Open
Abstract
Background Acute promyelocytic leukemia (APL) is a highly curable disease when treated with all-trans retinoid acid (ATRA) and arsenic trioxide (ATO). The combination of ATO and ATRA has become the standard therapeutic protocol for induction therapy in non-high-risk APL. An oral arsenic realgar-indigo naturalis formula (RIF) has also showed high efficacy and it has a more convenient route of administration than the standard intravenous regimen. Unlike in previous trials, the arsenical agent was used simultaneously with ATRA during post-remission therapy in this trial. Methods This study was designed as a multicenter, randomized controlled trial. The trial has a non-inferiority design with superiority being explored if non-inferiority is identified. All patients receive ATRA-ATO during the induction therapy. After achieving hematologic complete remission (HCR), patients were randomly assigned (1:1) to receive treatment with ATRA-RIF (experimental group) or ATRA-ATO (control group) as the consolidation therapy. During the consolidation therapy, the two groups receive ATRA plus RIF or intravenous ATO 2 weeks on and 2 to ~ 4 weeks off until molecular complete remission (MCR), then ATRA and oral RIF 2 weeks on and 2 to ~ 4 weeks off giving a total of six courses. Discussion This trial aims to compare the efficacy of ATRA-ATO versus ATRA-RIF in non-high-risk patients with APL, to demonstrate that oral RIF application reduces the total hospitalization days and medical costs. The simple schedule was studied in this trial. Trial registration ClinicalTrials.gov, NCT02899169. Registered on 14 September 2016.
Collapse
|
6
|
Basova S, Wilke N, Koch JC, Prokop A, Berkessel A, Pradel G, Ngwa CJ. Organoarsenic Compounds with In Vitro Activity against the Malaria Parasite Plasmodium falciparum. Biomedicines 2020; 8:biomedicines8080260. [PMID: 32748808 PMCID: PMC7459655 DOI: 10.3390/biomedicines8080260] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2020] [Revised: 07/24/2020] [Accepted: 07/29/2020] [Indexed: 01/16/2023] Open
Abstract
The rapid development of parasite drug resistance as well as the lack of medications targeting both the asexual and the sexual blood stages of the malaria parasite necessitate the search for novel antimalarial compounds. Eleven organoarsenic compounds were synthesized and tested for their effect on the asexual blood stages and sexual transmission stages of the malaria parasite Plasmodium falciparum using in vitro assays. The inhibitory potential of the compounds on blood stage viability was tested on the chloroquine (CQ)-sensitive 3D7 and the CQ-resistant Dd2 strain using the Malstat assay. The most effective compounds were subsequently investigated for their effect on impairing gametocyte development and gametogenesis, using the gametocyte-producing NF54 strain in respective cell-based assays. Their potential toxicity was investigated on leukemia cell line Nalm-6 and non-infected erythrocytes. Five out of the 11 compounds showed antiplasmodial activities against 3D7, with half-maximal inhibitory concentration (IC50) values ranging between 1.52 and 8.64 µM. Three of the compounds also acted against Dd2, with the most active compound As-8 exhibiting an IC50 of 0.35 µM. The five compounds also showed significant inhibitory effects on the parasite sexual stages at both IC50 and IC90 concentrations with As-8 displaying the best gametocytocidal activity. No hemolytic and cytotoxic effect was observed for any of the compounds. The organoarsenic compound As-8 may represent a good lead for the design of novel organoarsenic drugs with combined antimalarial and transmission blocking activities.
Collapse
Affiliation(s)
- Sofia Basova
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (S.B.); (G.P.)
| | - Nathalie Wilke
- Department of Paediatric Oncology, Children’s Hospital Cologne, Amsterdamer Straße 59, 50735 Cologne, Germany; (N.W.); (A.P.)
| | - Jan Christoph Koch
- Department of Chemistry, Organic Chemistry, University of Cologne, Greinstraße 4, 50939 Cologne, Germany; (J.C.K.); (A.B.)
| | - Aram Prokop
- Department of Paediatric Oncology, Children’s Hospital Cologne, Amsterdamer Straße 59, 50735 Cologne, Germany; (N.W.); (A.P.)
- Department of Paediatric Oncology, Helios Hospital Schwerin, Wismarsche Strasse 393-397, 19049 Schwerin, Germany
| | - Albrecht Berkessel
- Department of Chemistry, Organic Chemistry, University of Cologne, Greinstraße 4, 50939 Cologne, Germany; (J.C.K.); (A.B.)
| | - Gabriele Pradel
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (S.B.); (G.P.)
| | - Che Julius Ngwa
- Division of Cellular and Applied Infection Biology, Institute of Zoology, RWTH Aachen University, Worringerweg 1, 52074 Aachen, Germany; (S.B.); (G.P.)
- Correspondence:
| |
Collapse
|
7
|
Saad-El-Din AA, El-Tanahy ZH, El-Sayed SN, Anees LM, Farroh HA. Combined effect of arsenic trioxide and radiation on physical properties of hemoglobin biopolymer. JOURNAL OF RADIATION RESEARCH AND APPLIED SCIENCES 2019. [DOI: 10.1016/j.jrras.2014.07.005] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Aisha A. Saad-El-Din
- Biophys., Lab. Rad. Phys. Dep., National Center of Radiation Research and Technology (NCRRT), AEA, P.O. Box 29, Madinat Nasr, Cairo, Egypt
| | - Zinab H. El-Tanahy
- Nucl. Phys. Dep., Faculty of Science, Girls Branch, Al-Azhar University, Cairo, Egypt
| | - Suzan N. El-Sayed
- Solid Stat. Phys. Dep., Faculty of Science, Girls Branch, Al-Azhar University, Cairo, Egypt
| | - Laila M. Anees
- Health Res. Dep., National Center of Radiation Research and Technology (NCRRT), AEA, P.O. Box 29, Madinat Nasr, Cairo, Egypt
| | - Hoda A. Farroh
- Health Res. Dep., National Center of Radiation Research and Technology (NCRRT), AEA, P.O. Box 29, Madinat Nasr, Cairo, Egypt
| |
Collapse
|
8
|
Sheldon LA. Inhibition of E2F1 activity and cell cycle progression by arsenic via retinoblastoma protein. Cell Cycle 2017; 16:2058-2072. [PMID: 28880708 DOI: 10.1080/15384101.2017.1338221] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
The regulation of cell cycle progression by steroid hormones and growth factors is important for maintaining normal cellular processes including development and cell proliferation. Deregulated progression through the G1/S and G2/M cell cycle transitions can lead to uncontrolled cell proliferation and cancer. The transcription factor E2F1, a key cell cycle regulator, targets genes encoding proteins that regulate cell cycle progression through the G1/S transition as well as proteins important in DNA repair and apoptosis. E2F1 expression and activity is inhibited by inorganic arsenic (iAs) that has a dual role as a cancer therapeutic and as a toxin that leads to diseases including cancer. An understanding of what underlies this dichotomy will contribute to understanding how to use iAs as a more effective therapeutic and also how to treat cancers that iAs promotes. Here, we show that quiescent breast adenocarcinoma MCF-7 cells treated with 17-β estradiol (E2) progress through the cell cycle, but few cells treated with E2 + iAs progress from G1 into S-phase due to a block in cell cycle progression. Our data support a model in which iAs inhibits the dissociation of E2F1 from the tumor suppressor, retinoblastoma protein (pRB) due to changes in pRB phosphorylation which leads to decreased E2F1 transcriptional activity. These findings present an explanation for how iAs can disrupt cell cycle progression through E2F1-pRB and has implications for how iAs acts as a cancer therapeutic as well as how it may promote tumorigenesis through decreased DNA repair.
Collapse
Affiliation(s)
- Lynn A Sheldon
- a Geisel School of Medicine at Dartmouth, Department of Molecular and Systems Biology , Hanover , NH , USA
| |
Collapse
|
9
|
Abstract
Considerable debate exists regarding the potential antineoplastic effect of dietary long-chain n-3 PUFA contained in fatty fishes. Since the majority of published data has proven that their intake does not induce toxic or carcinogenic effects in humans, their possible preventive use against cancer has been suggested. On the other hand, it is unlikely that they could be effective in cancer patients as a single therapy. Nevertheless, a considerable effort has been put forth in recent years to evaluate the hypothesis that n-3 PUFA might improve the antineoplastic efficiency of currently used anticancer agents. The rationale for this therapeutic combinatory strategy is trying to increase cancer sensitivity to conventional therapies. This could allow the use of lower drug/radiation doses and, thereby, a reduction in the detrimental health effects associated with these treatments. We will here critically examine the studies that have investigated this possibility, by focusing particularly on the biological and molecular mechanisms underlying the antineoplastic effect of these combined treatments. A possible use of n-3 PUFA in combination with the innovative single-targeted anti-cancer therapies, that often are not completely devoid of dangerous side-effects, is also suggested.
Collapse
|
10
|
Eskandari F, Momeni HR. Silymarin protects plasma membrane and acrosome integrity in sperm treated with sodium arsenite. Int J Reprod Biomed 2016. [DOI: 10.29252/ijrm.14.1.47] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023] Open
|
11
|
Arsenic trioxide suppresses transcription of hTERT through down-regulation of multiple transcription factors in HL-60 leukemia cells. Toxicol Lett 2015; 232:481-9. [DOI: 10.1016/j.toxlet.2014.11.028] [Citation(s) in RCA: 21] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/28/2014] [Revised: 11/02/2014] [Accepted: 11/24/2014] [Indexed: 11/23/2022]
|
12
|
Song LL, Tu YY, Xia L, Wang WW, Wei W, Ma CM, Wen DH, Lei H, Xu HZ, Wu YL. Targeting catalase but not peroxiredoxins enhances arsenic trioxide-induced apoptosis in K562 cells. PLoS One 2014; 9:e104985. [PMID: 25115845 PMCID: PMC4130628 DOI: 10.1371/journal.pone.0104985] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/19/2014] [Accepted: 07/16/2014] [Indexed: 11/25/2022] Open
Abstract
Despite considerable efficacy of arsenic trioxide (As2O3) in acute promyelocytic leukemia (APL) treatment, other non-APL leukemias, such as chronic myeloid leukemia (CML), are less sensitive to As2O3 treatment. However, the underlying mechanism is not well understood. Here we show that relative As2O3-resistant K562 cells have significantly lower ROS levels than As2O3-sensitive NB4 cells. We compared the expression of several antioxidant enzymes in these two cell lines and found that peroxiredoxin 1/2/6 and catalase are expressed at high levels in K562 cells. We further investigated the possible role of peroxirdoxin 1/2/6 and catalase in determining the cellular sensitivity to As2O3. Interestingly, knockdown of peroxiredoxin 1/2/6 did not increase the susceptibility of K562 cells to As2O3. On the contrary, knockdown of catalase markedly enhanced As2O3-induced apoptosis. In addition, we provide evidence that overexpression of BCR/ABL cannot increase the expression of PRDX 1/2/6 and catalase. The current study reveals that the functional role of antioxidant enzymes is cellular context and treatment agents dependent; targeting catalase may represent a novel strategy to improve the efficacy of As2O3 in CML treatment.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Apoptosis/genetics
- Apoptosis/physiology
- Arsenic Trioxide
- Arsenicals/pharmacology
- Catalase/antagonists & inhibitors
- Catalase/genetics
- Catalase/metabolism
- Cell Line, Tumor
- Fusion Proteins, bcr-abl/genetics
- Fusion Proteins, bcr-abl/metabolism
- Gene Knockdown Techniques
- Humans
- K562 Cells
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/drug therapy
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/metabolism
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/pathology
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/metabolism
- Leukemia, Promyelocytic, Acute/pathology
- Oxides/pharmacology
- Peroxiredoxins/antagonists & inhibitors
- Peroxiredoxins/genetics
- Peroxiredoxins/metabolism
- Reactive Oxygen Species/metabolism
Collapse
Affiliation(s)
- Li-Li Song
- Department of Pathophysiology, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Yao-Yao Tu
- Department of Pathophysiology, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Li Xia
- Department of Pathophysiology, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei-Wei Wang
- Department of Pathophysiology, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Wei Wei
- Department of Pathophysiology, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Chun-Min Ma
- Department of Pathophysiology, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Dong-Hua Wen
- Department of Pathophysiology, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Hu Lei
- Department of Pathophysiology, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | - Han-Zhang Xu
- Department of Pathophysiology, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (HX); (YW)
| | - Ying-Li Wu
- Department of Pathophysiology, Chemical Biology Division of Shanghai Universities E-Institutes, Key Laboratory of Cell Differentiation and Apoptosis of the Chinese Ministry of Education, Shanghai Jiao Tong University School of Medicine, Shanghai, China
- * E-mail: (HX); (YW)
| |
Collapse
|
13
|
Abstract
It has been known for many years that arsenic trioxide (As 2O 3; ATO) is an effective therapy for acute promyelocytic leukemia but has little activity against other forms of the disease. ATO has diverse modes of action, but is well known to generate high levels of reactive oxygen species in cells which are believed to be causal in many of its biologic actions. ROS can both activate and suppress signaling through multiple intracellular pathways based on the amount and duration of ROS production. As the basal activity of the MEK1/2-ERK1/2 pathway is often high in acute myeloid leukemias, and that ATO is known to stimulate MEK1/2-ERK1/2 signaling in leukemia, the authors investigated whether knock down of the downstream effector of ERK1/2, RSK1, could enhance the anti-leukemic activity of ATO.
Collapse
Affiliation(s)
- Paul Dent
- Department of Neurosurgery; Massey Cancer Center; Virginia Commonwealth University; Richmond, VA USA
| |
Collapse
|
14
|
Salvianolic Acid B prevents arsenic trioxide-induced cardiotoxicity in vivo and enhances its anticancer activity in vitro. EVIDENCE-BASED COMPLEMENTARY AND ALTERNATIVE MEDICINE 2013; 2013:759483. [PMID: 23662152 PMCID: PMC3638618 DOI: 10.1155/2013/759483] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 12/22/2012] [Revised: 03/02/2013] [Accepted: 03/02/2013] [Indexed: 12/02/2022]
Abstract
Clinical attempts to reduce the cardiotoxicity of arsenic trioxide (ATO) without compromising its anticancer activities remain to be an unresolved issue. In this study, we determined whether Sal B can protect against ATO-induced cardiac toxicity in vivo and increase the toxicity of ATO toward cancer cells. Combination treatment of Sal B and ATO was investigated using BALB/c mice and human hepatoma (HepG2) cells and human cervical cancer (HeLa) cells. The results showed that the combination treatment significantly improved the ATO-induced loss of cardiac function, attenuated damage of cardiomyocytic structure, and suppressed the ATO-induced release of cardiac enzymes into serum in BALB/c mouse models. The expression levels of Bcl-2 and p-Akt in the mice treated with ATO alone were reduced, whereas those in the mice given the combination treatment were similar to those in the control mice. Moreover, the combination treatment significantly enhanced the ATO-induced cytotoxicity and apoptosis of HepG2 cells and HeLa cells. Increases in apoptotic marker cleaved poly (ADP-ribose) polymerase and decreases in procaspase-3 expressions were observed through western blot. Taken together, these observations indicate that the combination treatment of Sal B and ATO is potentially applicable for treating cancer with reduced cardiotoxic side effects.
Collapse
|
15
|
Abstract
The aim of this study was to investigate the hepatotoxicity induced by arsenic trioxide (As2O3) at a therapeutic dose for pediatric acute promyelocytic leukemia (APL). A total of APL patients received As2O3 treatment by IV drip. Hepatotoxicity was monitored based on the observations of the dynamic changes in liver function and treatment responses. The influencing factors of hepatotoxicity were further analyzed. Liver impairment occurred in 24.4% of the patients, most of which was mild and moderate in severity. Liver impairment was primarily manifested by increases in alanine aminotransferase, aspertate aminotransferase, and γ-glutamyl transferase, and these increases mostly occurred within 1 to 3 weeks and then returned to normal levels after 4 weeks. Patients' ages, sex, disease time, hepatomegaly and liver dysfunction, and disseminated intravascular coagulation did not show correlations with hepatotoxicity. Their hemogram and lactate dehydrogenase numerical values obtained from preliminary diagnosis did not show significant correlations with hepatotoxicity. Aggravated hepatotoxicity was not observed in patients receiving marcellomycin for the sake of complete remission. Changes in short-term cumulative dose of As2O3 did not exhibit a correlation with hepatotoxicity. However, patients suffering from differentiation syndrome were more likely at the risk of hepatotoxicity. Liver impairment improved after the suspension of As2O3 and liver protection treatment. No patient died of liver failure. Hepatotoxicity induced by As2O3 at a therapeutic dose for pediatric APL is mild and temporary. The first to the third week of the remission induction is the important period for monitoring. Great attention should be given to differentiation syndrome as it may worsen hepatotoxicity.
Collapse
|
16
|
Liu SL, He W, Wang RP, Li Z, Wu J, Zhao Y, Hu XH. Effects of recombinant hIL-2-Luffin P1 immunotoxin combining with arotinoid ethylester on the proliferation and apoptosis of Hut-78 cells. Mol Cell Toxicol 2013. [DOI: 10.1007/s13273-012-0041-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
|
17
|
Mancini I, Defant A. Bioactive Poly(Arsenic) Compounds. BIOMEDICAL INORGANIC POLYMERS 2013; 54:175-95. [DOI: 10.1007/978-3-642-41004-8_7] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/06/2023]
|
18
|
Zhang L, Sun H, Zhao F, Lu P, Ge C, Li H, Hou H, Yan M, Chen T, Jiang G, Xie H, Cui Y, Huang X, Fan J, Yao M, Li J. BMP4 administration induces differentiation of CD133+ hepatic cancer stem cells, blocking their contributions to hepatocellular carcinoma. Cancer Res 2012; 72:4276-85. [PMID: 22773665 DOI: 10.1158/0008-5472.can-12-1013] [Citation(s) in RCA: 78] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
CD133+ cancer stem cells (CSC) contribute to hepatocellular carcinoma (HCC) progression and resistance to therapy. Bone morphogenetic protein BMP4 plays an important role in hepatogenesis and hepatic stem cell differentiation, but little is known about its function in hepatic CSCs. In this study, we showed that high-dose exogenous BMP4 promotes CD133+ HCC CSC differentiation and inhibits the self-renewal, chemotherapeutic resistance, and tumorigenic capacity of these cells. Interestingly, we found that low-dose exogenous BMP4 upregulated CD133 protein expression in vitro, and endogenous BMP4 was preferentially expressed in CD133+ HCC CSCs, suggesting that low doses of BMP4 may facilitate CSC maintenance. A reduction in endogenous BMP4 levels decreased CD133 protein expression in vitro. In HCC tissues, expression of the BMP4 signaling target gene SMAD6 was positively correlated with CD133 expression. Activation of the Erk1/2 signaling pathway led to BMP4-mediated reduction in CD133 expression, which was reversed by treatment with MEK inhibitors. Taken together, our findings indicated that BMP4 might be a potent therapeutic agent in HCC that targets CSCs.
Collapse
Affiliation(s)
- Lixing Zhang
- State Key Laboratrory of Oncogenes and Related Genes, Shanghai Cancer Institute, Renji Hospital, Shanghai Jiaotong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Wang Y, Yang J, Yang K, Cang H, Huang XZ, Li H, Yi J. The biphasic redox sensing of SENP3 accounts for the HIF-1 transcriptional activity shift by oxidative stress. Acta Pharmacol Sin 2012; 33:953-63. [PMID: 22684029 DOI: 10.1038/aps.2012.40] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
AIM To investigate the mechanisms underlying the biphasic redox regulation of hypoxia-inducible factor-1 (HIF-1) transcriptional activity under different levels of oxidative stress caused by reactive oxidative species (ROS). METHODS HeLa cells were exposed to different concentrations of H(2)O(2) as a simple model for mild and severe oxidative stress. Luciferase reporter assay and/or quantitative real-time PCR were used to investigate the transcriptional activity. Immunoblot was used to detect protein expression. Chromatin immunoprecipitation assay was used to detect HIF-1/DNA binding. The interaction of p300 with HIF-1α or with SENP3, and the SUMO2/3 conjugation states of p300 were examined by coimmunoprecipitation. RESULTS HIF-1 transcriptional activity in HeLa cells was enhanced by low doses (0.05-0.5 mmol/L) of H(2)O(2), but suppressed by high doses (0.75-8.0 mmol/L) of H(2)O(2). The amount of co-activator p300 bound to HIF-1α in HeLa cells was increased under mild oxidative stress, but decreased under severe oxidative stress. The ROS levels differentially modified cysteines 243 and 532 in the cysteine protease SENP3, regulating the interaction of SENP3 with p300 to cause different SUMOylation of p300, thus shifting HIF-1 transcriptional activity. CONCLUSION The shift of HIF-1 transactivation by ROS is correlated with and dependent on the biphasic redox sensing of SENP3 that leads to the differential SENP3/p300 interaction and the consequent fluctuation in the p300 SUMOylation status.
Collapse
|
20
|
Momeni HR, Oryan S, Eskandari N. Effect of vitamin E on sperm number and testis histopathology of sodium arsenite-treated rats. Reprod Biol 2012; 12:171-81. [DOI: 10.1016/s1642-431x(12)60084-9] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
21
|
Bu R, Uddin S, Ahmed M, Hussain AR, Alsobhi S, Amin T, Al-Nuaim A, Al-Dayel F, Abubaker J, Bavi P, Al-Kuraya KS. c-Met inhibitor synergizes with tumor necrosis factor-related apoptosis-induced ligand to induce papillary thyroid carcinoma cell death. Mol Med 2012; 18:167-77. [PMID: 22113498 DOI: 10.2119/molmed.2011.00238] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/11/2011] [Accepted: 11/15/2011] [Indexed: 11/06/2022] Open
Abstract
The Met receptor tyrosine kinase is overexpressed and/or activated in variety of human malignancies. Previously we have shown that c-Met is overexpressed in Middle Eastern papillary thyroid carcinoma (PTC) and significantly associated with an aggressive phenotype, but its role has not been fully elucidated in PTC. The aim of this study was to determine the functional link between the c-Met/AKT signaling pathway and death receptor 5 (DR5) in a large cohort of PTC in a tissue microarray format followed by functional studies using PTC cell lines and nude mice. Our data showed that high expressions of p-Met and DR5 were significantly associated with an aggressive phenotype of PTC and correlated with BRAF mutation. Treatment of PTC cell lines with PHA665752, an inhibitor of c-Met tyrosine kinase, inhibited cell proliferation and induced apoptosis via the mitochondrial pathway in PTC cell lines. PHA665752 treatment or expression of c-Met small interfering (si)RNA resulted in dephosphorylation of c-Met, AKT and its downstream effector molecules. Furthermore, PHA665752 treatment upregulated DR5 expression via generation of reactive oxygen species in PTC cell lines, and synergistically potentiated death receptor-induced apoptosis with tumor necrosis factor-related apoptosis-inducing ligand (TRAIL). Finally, cotreatment with PHA665752 and TRAIL caused more pronounced effects on PTC xenograft tumor growth in nude mice. Our data suggest that the c-Met/AKT pathway may be a potential target for therapeutic intervention for treatment of PTC refractory to conventionally therapeutic modalities.
Collapse
Affiliation(s)
- Rong Bu
- Human Cancer Genomic Research, Research Center, Riyadh, Saudi Arabia
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Lombardo T, Cavaliere V, Costantino SN, Kornblihtt L, Alvarez EM, Blanco GA. Synergism between arsenite and proteasome inhibitor MG132 over cell death in myeloid leukaemic cells U937 and the induction of low levels of intracellular superoxide anion. Toxicol Appl Pharmacol 2011; 258:351-66. [PMID: 22178740 DOI: 10.1016/j.taap.2011.11.018] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2011] [Revised: 11/21/2011] [Accepted: 11/28/2011] [Indexed: 10/14/2022]
Abstract
Increased oxygen species production has often been cited as a mechanism determining synergism on cell death and growth inhibition effects of arsenic-combined drugs. However the net effect of drug combination may not be easily anticipated solely from available knowledge of drug-induced death mechanisms. We evaluated the combined effect of sodium arsenite with the proteasome inhibitor MG132, and the anti-leukaemic agent CAPE, on growth-inhibition and cell death effect in acute myeloid leukaemic cells U937 and Burkitt's lymphoma-derived Raji cells, by the Chou-Talalay method. In addition we explored the association of cytotoxic effect of drugs with changes in intracellular superoxide anion (O₂⁻) levels. Our results showed that combined arsenite+MG132 produced low levels of O₂⁻ at 6h and 24h after exposure and were synergic on cell death induction in U937 cells over the whole dose range, although the combination was antagonistic on growth inhibition effect. Exposure to a constant non-cytotoxic dose of 80μM hydrogen peroxide together with arsenite+MG132 changed synergism on cell death to antagonism at all effect levels while increasing O₂⁻ levels. Arsenite+hydrogen peroxide also resulted in antagonism with increased O₂⁻ levels in U937 cells. In Raji cells, arsenite+MG132 also produced low levels of O₂⁻ at 6h and 24h but resulted in antagonism on cell death and growth inhibition. By contrast, the combination arsenite+CAPE showed high levels of O₂⁻ production at 6h and 24 h post exposure but resulted in antagonism over cell death and growth inhibition effects in U937 and Raji cells. We conclude that synergism between arsenite and MG132 in U937 cells is negatively associated to O₂⁻ levels at early time points after exposure.
Collapse
Affiliation(s)
- Tomás Lombardo
- Laboratorio de Immunotoxicologia (LaITO), IDEHU-CONICET, Hospital de Clínicas, José de San Martín, Universidad de Buenos Aires (UBA), Buenos Aires, Argentina
| | | | | | | | | | | |
Collapse
|
23
|
Arsenic-based antineoplastic drugs and their mechanisms of action. Met Based Drugs 2011; 2008:260146. [PMID: 18431449 PMCID: PMC2292810 DOI: 10.1155/2008/260146] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2007] [Revised: 07/03/2007] [Accepted: 08/17/2007] [Indexed: 01/11/2023] Open
Abstract
Arsenic-based compounds have become accepted agents for cancer therapy providing high rates of remission of some cancers such as acute promyelocytic leukemia (APL). The mechanisms by which arsenic-containing compounds kill cells and
reasons for selective killing of only certain types of cancer cells such as APLs have recently been delineated. This knowledge
was gained in parallel with increasing understanding and awareness of the importance of intracellular redox systems and
regulation of the production of reactive oxygen species (ROS) by controlling mitochondrial function. Many of the targets for
the arsenic-containing compounds are mitochondrial proteins involved in regulating the production of ROS. Inhibition of these
proteins by disulfide linkage of vicinal thiol groups often leads to increased production of ROS and induction of apoptotic
signalling pathways. Sensitivity or resistance to the actions of arsenic-containing compounds on cancer cells and normal
cells depends on the levels of transport systems for their uptake or efflux from the cells as well as their redox defence
mechanisms. The exact mechanisms of arsenic toxicity as well as its anticancer properties are likely to be related and these
aspects of arsenic metabolism are covered in this review. Greater understanding of the mechanisms of action of arsenic will
help determine the risks of human exposure to this chemical. Novel organic arsenic-containing compounds and the lessons
learned from studying their selective sensitivity in targeting dividing endothelial cells to inhibit angiogenesis raise the future
possibility for designing better targeted antineoplastic arsenic-containing compounds with less toxicity to normal cells.
Collapse
|
24
|
Sardina JL, López-Ruano G, Sánchez-Sánchez B, Llanillo M, Hernández-Hernández A. Reactive oxygen species: are they important for haematopoiesis? Crit Rev Oncol Hematol 2011; 81:257-74. [PMID: 21507675 DOI: 10.1016/j.critrevonc.2011.03.005] [Citation(s) in RCA: 51] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2010] [Revised: 03/15/2011] [Accepted: 03/22/2011] [Indexed: 02/07/2023] Open
Abstract
The production of reactive oxygen species (ROS) has traditionally been related to deleterious effects for cells. However, it is now widely accepted that ROS can play an important role in regulating cellular signalling and gene expression. NADPH oxidase ROS production seems to be especially important in this regard. Some lines of evidence suggest that ROS may be important modulators of cell differentiation, including haematopoietic differentiation, in both physiologic and pathologic conditions. Here we shall review how ROS can regulate cell signalling and gene expression. We shall also focus on the importance of ROS for haematopoietic stem cell (HSC) biology and for haematopoietic differentiation. We shall review the involvement of ROS and NADPH oxidases in cancer, and in particular what is known about the relationship between ROS and haematological malignancies. Finally, we shall discuss the use of ROS as cancer therapeutic targets.
Collapse
Affiliation(s)
- José L Sardina
- Department of Biochemistry and Molecular Biology, University of Salamanca, Salamanca, Spain
| | | | | | | | | |
Collapse
|
25
|
Interactions of guanidinium with benzene-sulphonic, -phosphonic and -arsonic acids and several of their nitro-derivatives. J Mol Struct 2011. [DOI: 10.1016/j.molstruc.2010.11.062] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|
26
|
Salazar AM, Miller HL, McNeely SC, Sordo M, Ostrosky-Wegman P, States JC. Suppression of p53 and p21CIP1/WAF1 reduces arsenite-induced aneuploidy. Chem Res Toxicol 2010; 23:357-64. [PMID: 20000476 DOI: 10.1021/tx900353v] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Aneuploidy and extensive chromosomal rearrangements are common in human tumors. The role of DNA damage response proteins p53 and p21(CIP1/WAF1) in aneugenesis and clastogenesis was investigated in telomerase immortalized diploid human fibroblasts using siRNA suppression of p53 and p21(CIP1/WAF1). Cells were exposed to the environmental carcinogen sodium arsenite (15 and 20 microM), and the induction of micronuclei (MN) was evaluated in binucleated cells using the cytokinesis-block assay. To determine whether MN resulted from missegregation of chromosomes or from chromosomal fragments, we used a fluorescent in situ hybridization with a centromeric DNA probe. Micronuclei were predominantly of clastogenic origin in control cells regardless of p53 or p21(CIP1/WAF1) expression. MN with centromere signals in cells transfected with NSC siRNA or Mock increased 30% after arsenite exposure, indicating that arsenite induced aneuploidy in the tGM24 cells. Although suppression of p53 increased the fraction of arsenite-treated cells with MN, it caused a decrease in the fraction with centromeric DNA. Suppression of p21(CIP1/WAF1) like p53 suppression decreased the fraction of MN with centromeric DNA. Our results suggest that cells lacking normal p53 function cannot become aneuploid because they die by mitotic arrest-associated apoptosis, whereas cells with normal p53 function that are able to exit from mitotic arrest can become aneuploid. Furthermore, our current results support this role for p21(CIP1/WAF1) since suppression of p21(CIP1/WAF1) caused a decrease in aneuploidy induced by arsenite, suggesting that p21(CIP1/WAF1) plays a role in mitotic exit.
Collapse
Affiliation(s)
- Ana María Salazar
- Instituto de Investigaciones Biomedicas, Universidad Nacional Autonoma de Mexico, Mexico, D.F., Mexico
| | | | | | | | | | | |
Collapse
|
27
|
Mandegary A, Hosseini R, Ghaffari SH, Alimoghaddam K, Rostami S, Ghavamzadeh A, Ghahremani MH. The expression of p38, ERK1 and Bax proteins has increased during the treatment of newly diagnosed acute promyelocytic leukemia with arsenic trioxide. Ann Oncol 2010; 21:1884-1890. [PMID: 20164150 DOI: 10.1093/annonc/mdq034] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
BACKGROUND Promising reports exist regarding the use of arsenic trioxide (ATO) as first-line treatment in acute promyelocytic leukemia (APL). Although the in vitro effect of ATO is extensively studied, the in vivo mechanism(s) of ATO action is mostly unknown. PATIENTS AND METHODS Newly diagnosed APL patients were involved and received ATO (0.15 mg.kg/day) for 28 days as induction followed by consolidation therapy. Bone marrow (BM) aspirates were obtained on days 0, 14 and 28 of treatment for further molecular studies. Clinical findings and white blood cell counts were recorded as well. RESULTS Complete remission was observed in 17 (85%) patients with the median duration of 28 days (18-38) and cumulative dosage of median 280 mg (180-350). Hyperleukocytosis and APL differentiation syndrome (63%), gastrointestinal disorders (30%), liver enzyme elevation and night sweating (50%) were the most prevalent side-effects. The expression of Bax, ERK1 and p38 proteins and caspase-3 activity increased significantly in promyelocytes of BM aspirates at days 14 and 28 of induction therapy. CONCLUSION(S) These findings point toward the role of p38 and Bax in the induction of apoptosis, which was confirmed by increase in caspase-3 activity. However, the increase in ERK1 expression with regard to leukocytosis could translate to a proliferative/differentiation effect.
Collapse
Affiliation(s)
- A Mandegary
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran; Department of Pharmacology and Toxicology, Faculty of Pharmacy, Kerman University of Medical Sciences, Kerman
| | - R Hosseini
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran
| | - S H Ghaffari
- Hematology, Oncology and BMT Research Center, Tehran University of Medical Sciences, Shariati Hospital, Tehran
| | - K Alimoghaddam
- Hematology, Oncology and BMT Research Center, Tehran University of Medical Sciences, Shariati Hospital, Tehran; Department of Molecular Medicine, School of Advanced Medical Technologies, Tehran University of Medical Sciences, Tehran, Iran
| | - S Rostami
- Hematology, Oncology and BMT Research Center, Tehran University of Medical Sciences, Shariati Hospital, Tehran
| | - A Ghavamzadeh
- Hematology, Oncology and BMT Research Center, Tehran University of Medical Sciences, Shariati Hospital, Tehran
| | - M H Ghahremani
- Department of Pharmacology and Toxicology, Faculty of Pharmacy, Tehran University of Medical Sciences, Tehran; Department of Molecular Medicine, School of Advanced Medical Technologies, Tehran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
28
|
Ferrara F. Acute promyelocytic leukemia: what are the treatment options? Expert Opin Pharmacother 2010; 11:587-96. [DOI: 10.1517/14656560903505115] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
|
29
|
Manikam ST, Stanslas J. Andrographolide inhibits growth of acute promyelocytic leukaemia cells by inducing retinoic acid receptor-independent cell differentiation and apoptosis. J Pharm Pharmacol 2010. [DOI: 10.1211/jpp.61.01.0010] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
Abstract
Objectives
The growth inhibiting potential of andrographolide was evaluated in three acute promyelocytic leukaemia cell line models (HL-60, NB4 and all-trans retinoic acid (ATRA)-resistant NB4-R2).
Methods
In elucidating the mechanisms of growth inhibition, a special emphasis was placed on assessing the induction of differentiation and apoptosis by andrographolide in the primary acute promyelocytic leukaemia NB4 cells.
Key findings
The compound was 2- and 3-fold more active in inhibiting the growth of HL-60 and NB4-R2 cells compared with NB4 cells, respectively. At IC50 (concentration at which growth of 50% of the cells (compared with medium only treated control cells) is inhibited; 4.5 μM) the compound exhibited strong cell-differentiating activity in NB4 cells, similar to ATRA (IC50 1.5 μM). In the presence of a pure retinoic acid receptor antagonist AGN193109, the growth inhibition of NB4 cells by ATRA was reversed, whereas the activity of andrographolide was not affected. This clearly suggested that andrographolide's cell differentiating activity to induce growth inhibition of NB4 cells most likely occurred via a retinoic acid receptor-independent pathway. At higher concentration (2 × IC50), andrographolide was an efficient inducer of apoptosis in NB4 cells.
Conclusions
Taken together, these results suggest andrographolide and its derivatives, apparently with a novel cell differentiating mechanism and with ability to induce apoptosis, might be beneficial in the treatment of primary and ATRA-resistant acute promyelocytic leukaemia.
Collapse
Affiliation(s)
- Shiamala T Manikam
- Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| | - Johnson Stanslas
- Faculty of Medicine and Health Sciences, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
- Natural Products Laboratory, Institute of Bioscience, Universiti Putra Malaysia, Serdang, Selangor, Malaysia
| |
Collapse
|
30
|
Arsenic trioxide inhibits cholangiocarcinoma cell growth and induces apoptosis. Pathol Oncol Res 2009; 16:413-20. [PMID: 20012722 DOI: 10.1007/s12253-009-9234-1] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/16/2009] [Accepted: 11/19/2009] [Indexed: 12/18/2022]
Abstract
Arsenic trioxide (As(2)O(3)), an ingredient in many traditional Chinese medicines, has drawn broad attention due to its therapeutic effects on a variety of cancers, including some solid tumors. However, the effects of As(2)O(3) on cholangiocarcinoma have not been reported. In the present study, we demonstrate for the first time that clinically obtainable concentrations of As(2)O(3) inhibit cell growth and induce apoptosis in human cholangiocarcinoma SK-ChA-1 cells. As(2)O(3)-induced apoptosis was partially inhibited by caspase inhibitor and accompanied by changes in the expression of Bcl-2 family proteins, decrease of mitochondrial membrane potential (MMP), release of cytochrome C from mitochondria, activation of caspase-3, caspase-9, and cleavage of poly (ADP-ribose) polymerase (PARP). Thus As(2)O(3) induces apoptosis in SK-ChA-1 cells via mitochondria-mediated, caspases-dependent pathways. As(2)O(3) inhibition of Akt phosphorylation may contribute to As(2)O(3)-mediated cholangiocarcinoma cell growth inhibition and apoptosis induction.
Collapse
|
31
|
Preparation, characterization, in vivo and in vitro studies of arsenic trioxide Mg-Fe ferrite magnetic nanoparticles. Acta Pharmacol Sin 2009; 30:1688-93. [PMID: 19960013 DOI: 10.1038/aps.2009.158] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
AIM MgFe(2)O(4) magnetic nanoparticle composed of As(2)O(3) (As(2)O(3)-MNPs) were prepared and their in vitro and in vivo characteristics were studied. METHODS The solvent-displacement method was applied for preparation of the nanoparticle using Poly-D,L-lactic-co-glycolic acid(PLGA). The characteristics studies of the products included magnetic response, morphology (transmission electron microscopy and scanning electron microscopy), entrapment efficiency, drug loading, particle sizes, zeta potential, in vitro drug release and tissue magnetic targeting. Nanoparticle cytotoxicity to Saos-2 cells was investigated using the MTT assay. To guide the external magnetic field in the liver, the concentration of As(2)O(3) in the liver and kidney was measured using an atomic fluorescence spectrometer after injecting As(2)O(3)-MNPs into the caudal veins of mice. RESULTS The As(2)O(3)-MNPs were approximately spherical. The average diameter, drug loading, entrapment efficiency and zeta potential of As(2)O(3)-MNPs were 109.9 nm, 10.08%, 82.16%, and -14.33 mV, respectively. The specific saturation magnetism was 8.65 emu/g. In vivo, the concentration of As(2)O(3) in the liver was significantly higher than that in the non-magnetic group. While the concentration of As(2)O(3) in the kidney was lower than that in the non-magnetic group. The C(max) in liver tissue in the magnetic group was 30.65 microg/g, which was 4.17 times the drug concentration in the same group in kidney tissue (7.35 microg/g) and 2.88 times the concentration of drug (10.66 microg/g) in the liver tissue of the non-magnetic group. CONCLUSION The PLGA polymer-loaded magnetic nanoparticle composed of arsenic trioxide can be magnetically targeted well and applied in biomedicine.
Collapse
|
32
|
Bajorin DF, Halabi S, Small E. Arsenic Trioxide in Recurrent Urothelial Cancer: A Cancer and Leukemia Group B Phase II Trial (CALGB 99903). Clin Genitourin Cancer 2009; 7:E66-70. [DOI: 10.3816/cgc.2009.n.026] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
33
|
Chowdhury R, Chowdhury S, Roychoudhury P, Mandal C, Chaudhuri K. Arsenic induced apoptosis in malignant melanoma cells is enhanced by menadione through ROS generation, p38 signaling and p53 activation. Apoptosis 2009; 14:108-23. [PMID: 19082730 DOI: 10.1007/s10495-008-0284-8] [Citation(s) in RCA: 67] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
INTRODUCTION Resistance to apoptosis is a prominent feature of melanoma. Pharmacological concentration of arsenic in combination with a widely known oxidant, menadione was explored in this study to synergistically sensitize malignant melanoma cells to apoptosis. The molecular mechanism of apoptosis and the signaling-pathways involved were thoroughly investigated. MATERIALS METHODS AND RESULTS: Menadione synergized NaAsO(2) to significantly increase ROS generation and facilitate the major apoptotic signaling events: alteration of mitochondrial membrane potential, cytochrome c release and anti-apoptotic protein Bcl-2 down-regulation and subsequent activation of caspase-9 and caspase-3 followed by poly-ADP-ribose polymerase-1 cleavage. Antioxidant N-acetyl-L: -cysteine antagonized these events. Investigation of the signaling-pathway revealed significant suppression of AP-1 activity but not NF-kappaB upon NaAsO(2) and menadione application. An increase in p38 phosphorylation and p53 protein expression did also dictate the apoptotic response. Suppression of p38 activation with SB203580 and inhibition of p53 expression by siRNA attenuated apoptosis. Transfection of p53, in p53 null HCT cells augmented the apoptotic events. Moreover, the treatment also led to tumor size reduction in BALB/c mice developed by intra-dermal B16 mouse melanoma cell injection; however, it had no detectable pro-proliferative or pro-apoptotic effect on non-tumor keratinocytes, normal fibroblasts or PBMC. CONCLUSION This study thus provides an insight into innovative mechanisms of melanoma sensitization, a proper cure against which is still elusive. Taken together, our data also provides the first evidence of arsenic activity accentuation by menadione through modulation of specific signaling-pathways.
Collapse
Affiliation(s)
- Rajdeep Chowdhury
- Molecular & Human Genetics Division, Indian Institute of Chemical Biology, 4, Raja S.C. Mullick Road, Kolkata 700032, India
| | | | | | | | | |
Collapse
|
34
|
Wei L, Liao P, Wu H, Li X, Pei F, Li W, Wu Y. Metabolic profiling studies on the toxicological effects of realgar in rats by (1)H NMR spectroscopy. Toxicol Appl Pharmacol 2008; 234:314-25. [PMID: 19073202 DOI: 10.1016/j.taap.2008.11.010] [Citation(s) in RCA: 125] [Impact Index Per Article: 7.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/01/2008] [Revised: 11/10/2008] [Accepted: 11/10/2008] [Indexed: 01/07/2023]
Abstract
The toxicological effects of realgar after intragastrical administration (1 g/kg body weight) were investigated over a 21 day period in male Wistar rats using metabonomic analysis of (1)H NMR spectra of urine, serum and liver tissue aqueous extracts. Liver and kidney histopathology examination and serum clinical chemistry analyses were also performed. (1)H NMR spectra and pattern recognition analyses from realgar treated animals showed increased excretion of urinary Kreb's cycle intermediates, increased levels of ketone bodies in urine and serum, and decreased levels of hepatic glucose and glycogen, as well as hypoglycemia and hyperlipoidemia, suggesting the perturbation of energy metabolism. Elevated levels of choline containing metabolites and betaine in serum and liver tissue aqueous extracts and increased serum creatine indicated altered transmethylation. Decreased urinary levels of trimethylamine-N-oxide, phenylacetylglycine and hippurate suggested the effects on the gut microflora environment by realgar. Signs of impairment of amino acid metabolism were supported by increased hepatic glutamate levels, increased methionine and decreased alanine levels in serum, and hypertaurinuria. The observed increase in glutathione in liver tissue aqueous extracts could be a biomarker of realgar induced oxidative injury. Serum clinical chemistry analyses showed increased levels of lactate dehydrogenase, aspartate aminotransferase, and alkaline phosphatase as well as increased levels of blood urea nitrogen and creatinine, indicating slight liver and kidney injury. The time-dependent biochemical variations induced by realgar were achieved using pattern recognition methods. This work illustrated the high reliability of NMR-based metabonomic approach on the study of the biochemical effects induced by traditional Chinese medicine.
Collapse
Affiliation(s)
- Lai Wei
- Changchun Institute of Applied Chemistry, Chinese Academy of Sciences, Changchun, 130022, PR China
| | | | | | | | | | | | | |
Collapse
|
35
|
Li L, Wang J, Ye RD, Shi G, Jin H, Tang X, Yi J. PML/RARalpha fusion protein mediates the unique sensitivity to arsenic cytotoxicity in acute promyelocytic leukemia cells: Mechanisms involve the impairment of cAMP signaling and the aberrant regulation of NADPH oxidase. J Cell Physiol 2008; 217:486-93. [PMID: 18636556 DOI: 10.1002/jcp.21523] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Acute promyelocytic leukemia (APL) cells are characterized by PML/RARalpha fusion protein, high responsiveness to arsenic trioxide (ATO)-induced cytotoxicity and an abundant generation of reactive oxygen species (ROS). In this study we investigated the association among these three features in APL-derived NB4 cells. We found that NADPH oxidase-derived ROS generation was more abundant in NB4 cells compared with monocytic leukemia U937 cells. By using PR9, a sub-line of U937 stably transduced with the inducible PML/RARalpha expression vectors, we attributed disparities on ROS generation and ATO sensitivity to the occurrence of PML/RARalpha fusion protein, since PML/RARalpha-expressing cells appeared higher NADPH oxidase activity, higher ROS level and higher sensitivity to ATO. On the other hand, the basal intensity of cAMP signaling pathway was compared between NB4 and U937 as well as between PR9 cells with or without PML/RARalpha, demonstrating that PML/RARalpha-expressing cells had an impaired cAMP signaling pathway which relieved its inhibitory effect on NADPH oxidase derived ROS generation. In summary, the present study demonstrated the correlation of PML/RARalpha with cAMP signaling pathway, NADPH oxidase and ROS generation in APL cells. PML/RARalpha that bestows NB4 cells various pathological features, paradoxically also endows these cells with the basis for susceptibility to ATO-induced cytotoxcity.
Collapse
Affiliation(s)
- Lingna Li
- Department of Cell Biology, Key Laboratory of The Education Ministry for Cell Differentiation and Apoptosis, Institutes of Medical Sciencies, Shanghai Jiao Tong University School of Medicine, Shanghai, China
| | | | | | | | | | | | | |
Collapse
|
36
|
Liu J, Lu Y, Wu Q, Goyer RA, Waalkes MP. Mineral arsenicals in traditional medicines: orpiment, realgar, and arsenolite. J Pharmacol Exp Ther 2008; 326:363-8. [PMID: 18463319 PMCID: PMC2693900 DOI: 10.1124/jpet.108.139543] [Citation(s) in RCA: 135] [Impact Index Per Article: 7.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Mineral arsenicals have long been used in traditional medicines for various diseases, yet arsenic can be highly toxic and carcinogenic. Arsenic in traditional medicines typically comes from deliberate addition for therapeutic purposes, mainly in the form of mineral arsenicals, including orpiment (As2S3), realgar (As4S4), and arsenolite (contains arsenic trioxide, As2O3). Inorganic arsenic is now accepted in Western medicine as a first line chemotherapeutic agent against certain hematopoietic cancers. This perspective analyzes the pharmacology and toxicology of these arsenicals used in traditional medicines. Orpiment and realgar are less soluble and poorly absorbed from the gastrointestinal tract, whereas the bioavailability of arsenic trioxide is similar to inorganic arsenic salts such as sodium arsenite. Pharmacological studies show that arsenic trioxide and realgar are effective against certain malignancies. Orpiment and realgar are used externally for various skin diseases. Realgar is frequently included as an ingredient in oral traditional remedies for its antipyretic, anti-inflammatory, antiulcer, anti-convulsive, and anti-schistosomiasis actions, but the pharmacological basis for this inclusion still remains to be fully justified. Toxicological studies show that cardiovascular toxicity is the major concern for arsenic trioxide and that the gastrointestinal and dermal adverse effects may occur after prolonged use of mineral arsenicals. Little is known regarding the possible secondary cancers resulting from the long-term use of any of these arsenicals. Similar to the safety evaluation of seafood arsenicals, total arsenic content alone appears to be insufficient for mineral arsenical safety evaluation. Arsenic speciation, bioavailability, and toxicity/benefit should be considered in evaluation of mineral arsenical-containing traditional medicines.
Collapse
Affiliation(s)
- Jie Liu
- Inorganic Carcinogenesis Section, NCI at NIEHS, Mail Drop F0-09, Research Triangle Park, NC 27709, USA.
| | | | | | | | | |
Collapse
|
37
|
Phatak P, Dai F, Butler M, Nandakumar M, Gutierrez PL, Edelman MJ, Hendriks H, Burger AM. KML001 Cytotoxic Activity Is Associated with Its Binding to Telomeric Sequences and Telomere Erosion in Prostate Cancer Cells. Clin Cancer Res 2008; 14:4593-602. [DOI: 10.1158/1078-0432.ccr-07-4572] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
|
38
|
Munro KL, Mariana A, Klavins AI, Foster AJ, Lai B, Vogt S, Cai Z, Harris HH, Dillon CT. Microprobe XRF Mapping and XAS Investigations of the Intracellular Metabolism of Arsenic for Understanding Arsenic-Induced Toxicity. Chem Res Toxicol 2008; 21:1760-9. [DOI: 10.1021/tx800128d] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Kristie L. Munro
- School of Chemistry, University of Wollongong, NSW 2522, Australia, School of Chemistry, University of Sydney, NSW 2006, Australia, X-Ray Science Division, Argonne National Laboratory, Argonne, Illinois 60439, School of Chemistry and Physics, University of Adelaide, SA 5005, Australia
| | - Anna Mariana
- School of Chemistry, University of Wollongong, NSW 2522, Australia, School of Chemistry, University of Sydney, NSW 2006, Australia, X-Ray Science Division, Argonne National Laboratory, Argonne, Illinois 60439, School of Chemistry and Physics, University of Adelaide, SA 5005, Australia
| | - Andrejs I. Klavins
- School of Chemistry, University of Wollongong, NSW 2522, Australia, School of Chemistry, University of Sydney, NSW 2006, Australia, X-Ray Science Division, Argonne National Laboratory, Argonne, Illinois 60439, School of Chemistry and Physics, University of Adelaide, SA 5005, Australia
| | - Amalanie J. Foster
- School of Chemistry, University of Wollongong, NSW 2522, Australia, School of Chemistry, University of Sydney, NSW 2006, Australia, X-Ray Science Division, Argonne National Laboratory, Argonne, Illinois 60439, School of Chemistry and Physics, University of Adelaide, SA 5005, Australia
| | - Barry Lai
- School of Chemistry, University of Wollongong, NSW 2522, Australia, School of Chemistry, University of Sydney, NSW 2006, Australia, X-Ray Science Division, Argonne National Laboratory, Argonne, Illinois 60439, School of Chemistry and Physics, University of Adelaide, SA 5005, Australia
| | - Stefan Vogt
- School of Chemistry, University of Wollongong, NSW 2522, Australia, School of Chemistry, University of Sydney, NSW 2006, Australia, X-Ray Science Division, Argonne National Laboratory, Argonne, Illinois 60439, School of Chemistry and Physics, University of Adelaide, SA 5005, Australia
| | - ZhongHou Cai
- School of Chemistry, University of Wollongong, NSW 2522, Australia, School of Chemistry, University of Sydney, NSW 2006, Australia, X-Ray Science Division, Argonne National Laboratory, Argonne, Illinois 60439, School of Chemistry and Physics, University of Adelaide, SA 5005, Australia
| | - Hugh H. Harris
- School of Chemistry, University of Wollongong, NSW 2522, Australia, School of Chemistry, University of Sydney, NSW 2006, Australia, X-Ray Science Division, Argonne National Laboratory, Argonne, Illinois 60439, School of Chemistry and Physics, University of Adelaide, SA 5005, Australia
| | - Carolyn T. Dillon
- School of Chemistry, University of Wollongong, NSW 2522, Australia, School of Chemistry, University of Sydney, NSW 2006, Australia, X-Ray Science Division, Argonne National Laboratory, Argonne, Illinois 60439, School of Chemistry and Physics, University of Adelaide, SA 5005, Australia
| |
Collapse
|
39
|
Abstract
Research conducted over the last two decades has yielded a detailed understanding of the molecular lesions that contribute to the malignant transformation of hematopoietic stem cells and committed progenitors into the various forms of acute and chronic leukemia. Although our understanding of the molecular pathology of leukemia remains incomplete, the information gained to date has had a profound impact on the way these malignancies are both diagnosed and monitored during therapy. More recently, targeted therapies have been developed against some of the identified genetic lesions. These therapies have led to significant improvements in patient outcomes while simultaneously decreasing therapy-related toxicity. With the advent of genome-wide methods to define the total complement of genetic and epigenetic lesions involved in leukemogenesis, new targeted therapies can be anticipated. This review highlights some of the targeted therapies that are presently being used to treat hematopoietic malignancies and describes some of the recent advances that should have a significant impact on the development of future target therapies.
Collapse
Affiliation(s)
- James R Downing
- Department of Pathology, St Jude Children's Research Hospital, Memphis, TN 38105, USA.
| |
Collapse
|
40
|
Wang J, Li L, Cang H, Shi G, Yi J. NADPH oxidase-derived reactive oxygen species are responsible for the high susceptibility to arsenic cytotoxicity in acute promyelocytic leukemia cells. Leuk Res 2008; 32:429-36. [PMID: 17804067 DOI: 10.1016/j.leukres.2007.06.006] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2007] [Revised: 05/29/2007] [Accepted: 06/04/2007] [Indexed: 10/22/2022]
Abstract
We have previously demonstrated that an acute promyelocytic leukemia (APL)-derived cell line NB4 exhibited a relatively higher basal level of reactive oxygen species (ROS) than other leukemia cell lines, which is one of the mechanisms determining a higher apoptotic susceptibility of NB4 cells to arsenic trioxide (ATO)-induced apoptosis. Here we identified the source of the basal ROS generation in NB4 cells. We demonstrated the existence of all the components of phagocytic NADPH oxidase in NB4 cells and found that this oxidase could be effectively activated. The basal ROS generation in NB4 cells could be blocked by diphenyleneiodonium (DPI), an inhibitor of NADPH oxidase, but not by inhibitors of mitochondria respiratory chain, implying that NADPH oxidase played an essential role in maintaining the basal ROS level in NB4 cells. Furthermore, ATO-induced cytotoxicity was reduced by pre-treatment with DPI in NB4 cells, suggesting the involvement of NADPH oxidase in ATO-induced cytotoxicity. Therefore, increasing the NADPH oxidase activity may be a novel mechanism to enhance cytotoxicity induced by anticancer agents.
Collapse
Affiliation(s)
- Jie Wang
- Department of Cell Biology, Key Laboratory of The Education Ministry for Cell differentiation and Apoptosis, Shanghai Jiao Tong University School of Medicine, Shanghai 200025, China
| | | | | | | | | |
Collapse
|
41
|
Monomethylarsonate (MMAv) exerts stronger effects than arsenate on the structure and thermotropic properties of phospholipids bilayers. Biophys Chem 2008; 132:1-8. [DOI: 10.1016/j.bpc.2007.09.012] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2007] [Revised: 09/25/2007] [Accepted: 09/26/2007] [Indexed: 11/24/2022]
|
42
|
Liu T, Hannafon B, Gill L, Kelly W, Benbrook D. Flex-Hets differentially induce apoptosis in cancer over normal cells by directly targeting mitochondria. Mol Cancer Ther 2007; 6:1814-22. [PMID: 17575110 PMCID: PMC2701109 DOI: 10.1158/1535-7163.mct-06-0279] [Citation(s) in RCA: 77] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Flex-Het drugs induce apoptosis in multiple types of cancer cells, with little effect on normal cells. This apoptosis occurs through the intrinsic mitochondrial pathway accompanied by generation of reactive oxygen species (ROS). The objective of this study was to determine if direct or indirect targeting of mitochondria is responsible for the differential sensitivities of cancer and normal cells to Flex-Hets. Mitochondrial effects and apoptosis were measured using JC-1 and Annexin V-FITC dyes with flow cytometry. Bcl-2, Bcl-x(L), and Bax were measured by Western blot. Flex-Hets induced mitochondrial swelling and apoptosis in ovarian cancer cell lines but had minimal to no effects in a variety of normal cell cultures, including human ovarian surface epithelium. Effects on inner mitochondrial membrane (IMM) potential were variable and did not occur in normal cells. Two different antioxidants, administered at concentrations shown to quench intracellular and mitochondrial ROS, did not alter Flex-Het-induced mitochondrial swelling, loss of IMM potential, or apoptosis. Inhibition of protein synthesis with cycloheximide also did not prevent Flex-Het mitochondrial or apoptosis effects. Bcl-2 and Bcl-x(L) levels were decreased in an ovarian cancer cell line but increased in a normal culture, whereas Bax expression was unaffected by Flex-Hets treatment. In conclusion, ROS seems to be a consequence rather than a cause of mitochondrial swelling. The differential induction of apoptosis in cancer versus normal cells by Flex-Hets involves direct targeting of mitochondria associated with alterations in the balance of Bcl-2 proteins. This mechanism does not require IMM potential, ROS generation, or protein synthesis.
Collapse
Affiliation(s)
- Tongzu Liu
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Zhongnan Hospital, Wuhan University, Wuhan, Hubei, China
| | - Bethany Hannafon
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| | - Lance Gill
- Department of Chemistry, Southwestern Oklahoma State University, Weatherford, Oklahoma
| | - William Kelly
- Department of Chemistry, Southwestern Oklahoma State University, Weatherford, Oklahoma
| | - Doris Benbrook
- Department of Obstetrics and Gynecology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
- Department of Biochemistry and Molecular Biology, University of Oklahoma Health Sciences Center, Oklahoma City, Oklahoma
| |
Collapse
|
43
|
Dilda PJ, Hogg PJ. Arsenical-based cancer drugs. Cancer Treat Rev 2007; 33:542-64. [PMID: 17624680 DOI: 10.1016/j.ctrv.2007.05.001] [Citation(s) in RCA: 281] [Impact Index Per Article: 15.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/10/2007] [Revised: 05/21/2007] [Accepted: 05/23/2007] [Indexed: 01/04/2023]
Abstract
Arsenic is a semi-metal or metalloid with two biologically important oxidation states, As(III) and As(V). As(III), in particular, reacts with closely spaced protein thiols, forming stable cyclic dithioarsinite complexes in which both sulfur atoms are bound to arsenic. It is this reaction that is mostly responsible for arsenics cytotoxicity. Arsenic compounds have been used as medicinal agents for many centuries for the treatment of diseases such as psoriasis, syphilis, and rheumatosis. From the 1700's until the introduction of and use of modern chemotherapy and radiation therapy in the mid 1900's, arsenic was a mainstay in the treatment of leukemia. Concerns about the toxicity of arsenical compounds led eventually to their abandonment for the treatment of cancer. The discovery in the 1980's that arsenic trioxide induces complete remission in a high percentage of patients with acute promyelocytic leukemia has awakened interest in this metalloid for the treatment of human disease. In particular, a new class or organoarsenicals are being trialed for the treatment of hematological malignancies and solid tumors. In this review, we discuss the arsenical-based compounds used in the past and present for the treatment of various forms of cancer. Mechanisms of action and selectivity and acute and chronic toxicities are discussed along with the prospects of this class of molecule.
Collapse
Affiliation(s)
- Pierre J Dilda
- UNSW Cancer Research Centre, University of New South Wales and Department of Haematology, Prince of Wales Hospital, Sydney 2052, Australia
| | | |
Collapse
|
44
|
Hussain AR, Al-Jomah NA, Siraj AK, Manogaran P, Al-Hussein K, Abubaker J, Platanias LC, Al-Kuraya KS, Uddin S. Sanguinarine-dependent induction of apoptosis in primary effusion lymphoma cells. Cancer Res 2007; 67:3888-97. [PMID: 17440103 DOI: 10.1158/0008-5472.can-06-3764] [Citation(s) in RCA: 100] [Impact Index Per Article: 5.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Primary effusion lymphoma (PEL) is an incurable, aggressive B-cell malignancy that develops rapid resistance to conventional chemotherapy. In efforts to identify novel approaches to block proliferation of PEL cells, we found that sanguinarine, a natural compound isolated from the root plant Sanguinaria canadendid, inhibits cell proliferation and induces apoptosis in a dose-dependent manner in several PEL cell lines. Our data show that sanguinarine treatment of PEL cells results in up-regulation of death receptor 5 (DR5) expression via generation of reactive oxygen species (ROS) and causes activation of caspase-8 and truncation of Bid (tBid). Subsequently, tBid translocates to the mitochondria causing conformational changes in Bax, leading to loss of mitochondrial membrane potential and release of cytochrome c to the cytosol. Sanguinarine-induced release of cytochrome c results in activation of caspase-9 and caspase-3 and poly(ADP-ribose) polymerase (PARP) cleavage, leading to induction of caspase-dependent apoptosis. In addition, we show that pretreatment of PEL cells with carbobenzoxy-Val-Ala-Asp-fluoromethylketone, a universal inhibitor of caspases, abrogates caspase and PARP activation and prevents cell death induced by sanguinarine. Moreover, treatment of PEL cells with sanguinarine down-regulates expression of inhibitor of apoptosis proteins (IAP). Finally, N-acetylcysteine, an inhibitor of ROS, inhibits sanguinarine-induced generation of ROS, up-regulation of DR5, Bax conformational changes, activation of caspase-3, and down-regulation of IAPs. Taken together, our findings suggest that sanguinarine is a potent inducer of apoptosis of PEL cells via up-regulation of DR5 and raise the possibility that this agent may be of value in the development of novel therapeutic approaches for the treatment of PEL.
Collapse
Affiliation(s)
- Azhar R Hussain
- Human Cancer Genomic Research, King Fahad National Center for Children's Cancer and Research, Biological and Medical Research, Riyadh 11211, Saudi Arabia
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Pelicano H, Carew JS, McQueen TJ, Andreeff M, Plunkett W, Keating MJ, Huang P. Targeting Hsp90 by 17-AAG in leukemia cells: mechanisms for synergistic and antagonistic drug combinations with arsenic trioxide and Ara-C. Leukemia 2006; 20:610-9. [PMID: 16482209 DOI: 10.1038/sj.leu.2404140] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
17-Allylamino-17-demethoxygeldanamycin (17-AAG) is a new anticancer agent currently in clinical trials. The ability of 17-AAG to abrogate the function of heat-shock protein Hsp90 and modulate cellular sensitivity to anticancer agents has prompted recent research to use this compound in drug combination therapy. Here we report that 17-AAG has striking opposite effects on the activity of arsenic trioxide (ATO) and ara-C. Combination of 17-AAG with ATO exhibited a synergistic effect in leukemia cells, whereas coincubation of 17-AAG and ara-C showed antagonistic activity. Mechanistic studies revealed that ATO exerted cytotoxic action by reactive oxygen species generation, and activated Akt survival pathway. 17-AAG abrogated Akt activation and enhanced the activity of ATO. In contrast, treatment of leukemia cells with 17-AAG caused a G1 arrest, a decrease in DNA synthesis and reduced ara-C incorporation into DNA, leading to antagonism. The ability of 17-AAG to enhance the antileukemia activity of ATO was further demonstrated in primary leukemia cells isolated from patients with acute myeloid leukemia and chronic lymphocytic leukemia, including cells from refractory patients. Our data suggest that combination of 17-AAG and ATO may be an effective therapeutic regimen. Caution should be exercised in using 17-AAG together with ara-C, as their combination effects are schedule dependent.
Collapse
MESH Headings
- Acute Disease
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Arsenic Trioxide
- Arsenicals/antagonists & inhibitors
- Arsenicals/pharmacology
- Benzoquinones/pharmacology
- Cytarabine/antagonists & inhibitors
- Cytarabine/pharmacology
- DNA/biosynthesis
- DNA/drug effects
- DNA/metabolism
- Drug Administration Schedule
- Drug Antagonism
- Drug Screening Assays, Antitumor
- Drug Synergism
- Drug Therapy, Combination
- G1 Phase/drug effects
- HSP90 Heat-Shock Proteins/antagonists & inhibitors
- HSP90 Heat-Shock Proteins/physiology
- HeLa Cells
- Humans
- Jurkat Cells
- Lactams, Macrocyclic/pharmacology
- Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy
- Leukemia, Lymphocytic, Chronic, B-Cell/metabolism
- Leukemia, Myeloid/drug therapy
- Leukemia, Myeloid/metabolism
- Oxides/antagonists & inhibitors
- Oxides/pharmacology
- Sensitivity and Specificity
- Structure-Activity Relationship
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- H Pelicano
- Department of Molecular Pathology, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | | | |
Collapse
|
46
|
Abstract
Anion exchanger 2 (AE2) mediates the exchange of C1-/HCO3- across the plasma membrane and plays a role in the regulation of intracellular pH. The present study showed that AE2 protein expression was upregulated immediately after exposure to either low (0.5 micromol/l) or high (1 and 2 micromol/l) concentrations of arsenic trioxide. This suggests that arsenic trioxide may act via regulation of intracellular pH. Changing the culture pH in NB4 cells modulated the degradation of promyelocytic leukaemia-retinoic acid receptor-alpha (PML-RARalpha), PML and RARalpha, which supported this hypothesis. DIDS (4,4'-diisothiocyanodihydrostilbene-2,2'-disulphonic acid) inhibited AE2 function, preventing the arsenic trioxide-induced degradation of RARalpha and low concentration showed synergistic effects on the expression of CD11c, which is related with cell differentiation. In addition, DIDS rescued the cells from 1 micromol/l arsenic trioxide-induced apoptosis. In conclusion, AE2 mediated the action of arsenic trioxide via regulation of intracellular pH and a novel pathway for the mechanism of action of arsenic trioxide is reported.
Collapse
Affiliation(s)
- Xiao-Yan Pan
- Department of Pathophysiology, Shanghai Jiao Tong University, School of Medicine
| | | | | | | | | |
Collapse
|
47
|
Ge Y, Montano I, Rustici G, Freebern WJ, Haggerty CM, Cui W, Ponciano-Jackson D, Chandramouli GVR, Gardner ER, Figg WD, Abu-Asab M, Tsokos M, Jackson SH, Gardner K. Selective leukemic-cell killing by a novel functional class of thalidomide analogs. Blood 2006; 108:4126-35. [PMID: 16940421 PMCID: PMC1895447 DOI: 10.1182/blood-2006-04-017046] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/26/2023] Open
Abstract
Using a novel cell-based assay to profile transcriptional pathway targeting, we have identified a new functional class of thalidomide analogs with distinct and selective antileukemic activity. These agents activate nuclear factor of activated T cells (NFAT) transcriptional pathways while simultaneously repressing nuclear factor-kappaB (NF-kappaB) via a rapid intracellular amplification of reactive oxygen species (ROS). The elevated ROS is associated with increased intracellular free calcium, rapid dissipation of the mitochondrial membrane potential, disrupted mitochondrial structure, and caspase-independent cell death. This cytotoxicity is highly selective for transformed lymphoid cells, is reversed by free radical scavengers, synergizes with the antileukemic activity of other redox-directed compounds, and preferentially targets cells in the S phase of the cell cycle. Live-cell imaging reveals a rapid drug-induced burst of ROS originating in the endoplasmic reticulum and associated mitochondria just prior to spreading throughout the cell. As members of a novel functional class of "redoxreactive" thalidomides, these compounds provide a new tool through which selective cellular properties of redox status and intracellular bioactivation can be leveraged by rational combinatorial therapeutic strategies and appropriate drug design to exploit cell-specific vulnerabilities for maximum drug efficacy.
Collapse
Affiliation(s)
- Yun Ge
- The Advanced Technology Center, Laboratory of Receptor Biology and Gene Expression, National Cancer Institute, Bethesda, MD 20892-4605, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
48
|
Abstract
A wide array of recurrent, non-random chromosomal translocations are associated with hematologic malignancies; experimental models have clearly demonstrated that many of these translocations are causal events during malignant transformation. Translocations involving the MLL gene are among the most common of these non-random translocations. Leukemias with MLL translocations have been the topic of intense interest because of the unusual, biphenotypic immunophenotype of these leukemias, because of the unique clinical presentation of some MLL translocations (infant leukemia and therapy-related leukemia), and because of the large number of different chromosomal loci that partner with MLL in these translocations. This review is focused on the potential mechanisms that lead to MLL translocations, and will discuss aberrant VDJ recombination, Alu-mediated recombination, non-homologous end joining, as well as the effect of DNA topoisomerase II poisons and chromatin structure.
Collapse
Affiliation(s)
- Peter D Aplan
- Genetics Branch, Center for Cancer Research, National Cancer Institute, National Institutes of Health, National Naval Medical Center, Bldg. 8 Rm. 5101, 8901 Rockville Pike, Bethesda, MD 20889, USA.
| |
Collapse
|
49
|
Hornhardt S, Gomolka M, Walsh L, Jung T. Comparative investigations of sodium arsenite, arsenic trioxide and cadmium sulphate in combination with gamma-radiation on apoptosis, micronuclei induction and DNA damage in a human lymphoblastoid cell line. Mutat Res 2006; 600:165-76. [PMID: 16764896 DOI: 10.1016/j.mrfmmm.2006.04.002] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2006] [Revised: 04/04/2006] [Accepted: 04/13/2006] [Indexed: 10/24/2022]
Abstract
In the field of radiation protection the combined exposure to radiation and other toxic agents is recognised as an important research area. To elucidate the basic mechanisms of simultaneous exposure, the interaction of the carcinogens and environmental toxicants cadmium and two arsenic compounds, arsenite and arsenic trioxide, in combination with gamma-radiation in human lymphoblastoid cells (TK6) were investigated. Gamma-radiation induced significant genotoxic effects such as micronuclei formation, DNA damage and apoptosis, whereas arsenic and cadmium had no significant effect on these indicators of cellular damage at non-toxic concentrations. However, in combination with gamma-radiation arsenic trioxide induced a more than additive apoptotic rate compared to the sum of the single effects. Here, the level of apoptotic cells was increased, in a dose-dependent way, up to two-fold compared to the irradiated control cells. Arsenite did not induce a significant additive effect at any of the concentrations or radiation doses tested. On the other hand, arsenic trioxide was less effective than arsenite in the induction of DNA protein cross-links. These data indicate that the two arsenic compounds interact through different pathways in the cell. Cadmium sulphate, like arsenite, had no significant effect on apoptosis in combination with gamma-radiation at low concentrations and, at high concentrations, even reduced the radiation-induced apoptosis. An additive effect on micronuclei induction was observed with 1muM cadmium sulphate with an increase of up to 80% compared to the irradiated control cells. Toxic concentrations of cadmium and arsenic trioxide seemed to reduce micronuclei induction. The results presented here indicate that relatively low concentrations of arsenic and cadmium, close to those occuring in nature, may interfere with radiation effects. Differences in action of the two arsenic compounds were identified.
Collapse
Affiliation(s)
- Sabine Hornhardt
- BfS-Federal Office for Radiation Protection, Department of Radiation Protection and Health, Ingolstädter Landstr. 1, 85764 Oberschleissheim, Germany.
| | | | | | | |
Collapse
|
50
|
Gloire G, Legrand-Poels S, Piette J. NF-kappaB activation by reactive oxygen species: fifteen years later. Biochem Pharmacol 2006; 72:1493-505. [PMID: 16723122 DOI: 10.1016/j.bcp.2006.04.011] [Citation(s) in RCA: 1174] [Impact Index Per Article: 61.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2006] [Revised: 04/05/2006] [Accepted: 04/07/2006] [Indexed: 02/06/2023]
Abstract
The transcription factor NF-kappaB plays a major role in coordinating innate and adaptative immunity, cellular proliferation, apoptosis and development. Since the discovery in 1991 that NF-kappaB may be activated by H(2)O(2), several laboratories have put a considerable effort into dissecting the molecular mechanisms underlying this activation. Whereas early studies revealed an atypical mechanism of activation, leading to IkappaBalpha Y42 phosphorylation independently of IkappaB kinase (IKK), recent findings suggest that H(2)O(2) activates NF-kappaB mainly through the classical IKK-dependent pathway. The molecular mechanisms leading to IKK activation are, however, cell-type specific and will be presented here. In this review, we also describe the effect of other ROS (HOCl and (1)O(2)) and reactive nitrogen species on NF-kappaB activation. Finally, we critically review the recent data highlighting the role of ROS in NF-kappaB activation by proinflammatory cytokines (TNF-alpha and IL-1beta) and lipopolysaccharide (LPS), two major components of innate immunity.
Collapse
Affiliation(s)
- Geoffrey Gloire
- Center for Biomedical Integrated Genoproteomics (CBIG), Virology and Immunology Unit, University of Liège, 4000 Liège, Belgium
| | | | | |
Collapse
|