1
|
Alimohammadi M, Rahimzadeh P, Khorrami R, Bonyadi M, Daneshi S, Nabavi N, Raesi R, Farani MR, Dehkhoda F, Taheriazam A, Hashemi M. A comprehensive review of the PTEN/PI3K/Akt axis in multiple myeloma: From molecular interactions to potential therapeutic targets. Pathol Res Pract 2024; 260:155401. [PMID: 38936094 DOI: 10.1016/j.prp.2024.155401] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/04/2024] [Revised: 06/02/2024] [Accepted: 06/09/2024] [Indexed: 06/29/2024]
Abstract
Phosphatase and tensin homolog (PTEN), phosphatidylinositol 3-kinase (PI3K), and protein kinase B (Akt) signaling pathways contribute to the development of several cancers, including multiple myeloma (MM). PTEN is a tumor suppressor that influences the PI3K/Akt/mTOR pathway, which in turn impacts vital cellular processes like growth, survival, and treatment resistance. The current study aims to present the role of PTEN and PI3K/Akt/mTOR signaling in the development of MM and its response to treatment. In addition, the molecular interactions in MM that underpin the PI3K/Akt/mTOR pathway and address potential implications for the development of successful treatment plans are also discussed in detail. We investigate their relationship to both upstream and downstream regulators, highlighting new developments in combined therapies that target the PTEN/PI3K/Akt axis to overcome drug resistance, including the use of PI3K and mitogen-activated protein kinase (MAPK) inhibitors. We also emphasize that PTEN/PI3K/Akt pathway elements may be used in MM diagnosis, prognosis, and therapeutic targets.
Collapse
Affiliation(s)
- Mina Alimohammadi
- Department of Immunology, School of Medicine, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran.
| | - Payman Rahimzadeh
- Surgical Research Society (SRS), Students' Scientific Research Center, Tehran University of Medical Sciences, Tehran, Iran
| | - Ramin Khorrami
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Islamic Republic of Iran
| | - Mojtaba Bonyadi
- Department of Food Hygiene and Quality Control, Faculty of Veterinary Medicine, University of Tehran, Tehran, Islamic Republic of Iran
| | - Salman Daneshi
- Department of Public Health, School of Health, Jiroft University of Medical Sciences, Jiroft, Islamic Republic of Iran
| | - Noushin Nabavi
- Independent Researcher, Victoria, British Columbia V8V 1P7, Canada
| | - Rasoul Raesi
- Department of Health Services Management, Mashhad University of Medical Sciences, Mashhad, Islamic Republic of Iran; Department of Nursing, Torbat Jam Faculty of Medical Sciences, Torbat Jam, Iran
| | - Marzieh Ramezani Farani
- NanoBio High-Tech Materials Research Center, Department of Biological Sciences and Bioengineering, Inha University, 100 Inha-ro, Incheon 22212, Republic of Korea
| | - Farshid Dehkhoda
- Department of Orthopedics, Shahid Beheshti University of Medical Sciences, Tehran, Islamic Republic of Iran.
| | - Afshin Taheriazam
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Islamic Republic of Iran; Department of Orthopedics, Faculty of Medicine, Tehran Medical Sciences, Islamic Azad University, Tehran, Islamic Republic of Iran.
| | - Mehrdad Hashemi
- Farhikhtegan Medical Convergence Sciences Research Center, Farhikhtegan Hospital Tehran Medical Sciences, Islamic Azad University, Tehran, Islamic Republic of Iran; Department of Genetics, Faculty of Advanced Science and Technology, Tehran Medical Sciences, Islamic Azad University, Tehran, Islamic Republic of Iran.
| |
Collapse
|
2
|
Chen F, Zhong X, Dai Q, Li K, Zhang W, Wang J, Zhao Y, Shen J, Xiao Z, Xing H, Li J. Human Umbilical Cord MSC Delivered-Soluble TRAIL Inhibits the Proliferation and Promotes Apoptosis of B-ALL Cell In Vitro and In Vivo. Pharmaceuticals (Basel) 2022; 15:1391. [PMID: 36422522 PMCID: PMC9693801 DOI: 10.3390/ph15111391] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2022] [Revised: 11/09/2022] [Accepted: 11/09/2022] [Indexed: 09/19/2023] Open
Abstract
The TNF-related apoptosis-inducing ligand (TRAIL) could induce apoptosis of leukemic cells, while showed no cytotoxic effect on normal cells. One of the limitations for application of recombinant TRAIL (rhTRAIL) in leukemia treatment is that the serum half-life of this protein is short. Gene delivery is a good strategy to prolong the half-life of TRAIL. In this study, we genetically engineered umbilical cord-MSCs to continuously express and secrete soluble TRAIL (MSC-sTRAIL), to investigate the effects of MSC-sTRAIL on B-cell acute lymphocytic leukemia (B-ALL) cells. In vitro, MSC-sTRAIL significantly inhibited the proliferation of B-ALL cells by suppressing PI3K/AKT and MEK/ERK signaling pathways, and induced apoptosis of B-ALL cells via the caspase cascade-mediated pathway and mitochondrial-mediated pathway. In vivo, MSC-sTRAIL dramatically inhibited B-ALL cell growth. Meanwhile, B-ALL-induced splenic and renal injuries were significantly alleviated after MSC-sTRAIL treatment. Moreover, the serum levels of MSC-secreted sTRAIL were still high in MSC-sTRAIL treated mice, indicating an extended half-life of sTRAIL. Our study suggests that MSC delivered-TRAIL secretion is a potential therapeutic strategy for B-ALL treatment.
Collapse
Affiliation(s)
- Fangshan Chen
- Department of Oncology and Hematology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Xianmei Zhong
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
- Department of Pharmacy, People’s Hospital of Nanbu County, Nanchong 637300, China
| | - Qian Dai
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Kuo Li
- Department of Oncology and Hematology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Wei Zhang
- Department of Oncology and Hematology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Jie Wang
- Department of Oncology and Hematology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| | - Yueshui Zhao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Jing Shen
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Zhangang Xiao
- Laboratory of Molecular Pharmacology, Department of Pharmacology, School of Pharmacy, Southwest Medical University, Luzhou 646000, China
- Cell Therapy & Cell Drugs of Luzhou Key Laboratory, Luzhou 646000, China
- South Sichuan Institute of Translational Medicine, Luzhou 646000, China
| | - Hongyun Xing
- Department of Hematology, The Affiliated Hospital of Southwest Medical University, Luzhou 646000, China
| | - Jing Li
- Department of Oncology and Hematology, The Affiliated Traditional Chinese Medicine Hospital of Southwest Medical University, Luzhou 646000, China
| |
Collapse
|
3
|
Protective activity of tert-butylhydroquinone against oxidative stress and apoptosis induced by glutamate agonizts in R28 cells and mice retina. Biomed Pharmacother 2022; 152:113117. [PMID: 35653886 DOI: 10.1016/j.biopha.2022.113117] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2022] [Revised: 05/02/2022] [Accepted: 05/10/2022] [Indexed: 11/22/2022] Open
Abstract
Glutamate excitotoxicity can cause cell damage and apoptosis and play an important role in a variety of retinal diseases. Tertiary-butylhydroquinone (tBHQ) is an approved food-grade phenolic antioxidant with antioxidant activity in a variety of cells and tissues. We observed the protective effect of tBHQ on glutamatergic agonist-induced retina and explored its possible mechanism of action through in vitro cell experiments. The results showed that tBHQ had protective effects on NMDA-induced mouse retinal excitotoxicity and glutamate-induced excitotoxicity in rat retinal precursor cells (R28 cells). tBHQ reversed glutamate-induced apoptosis, production of intracellular reactive oxygen species, and reduction of mitochondrial membrane potential. Western blot analysis showed that tBHQ could increase the expression of procaspase-3, Bcl-2, AIF precursor, CAT, SOD2, Nrf2, NQO1, HO-1 and NF-κB in glutamate-treated cells, and decrease the expression of AIF cleavage products. Furthermore, we discovered that tBHQ activated müller glial cells. Based on these results, tBHQ may have antioxidant and anti-apoptotic properties, thus serving as a potential retinal protective agent. Its anti-oxidative stress effect was attributed to up-regulation of Nrf2, and its anti-apoptotic effect was related to its up-regulation of Bcl-2 expression and inhibition of mitochondria-dependent apoptosis.
Collapse
|
4
|
Abstract
STUDY DESIGN In vitro experimental study. OBJECTIVE To investigate the impact of increased osteoblastic activity on the proliferation and survival of multiple myeloma (MM) plasma cells in vitro SUMMARY OF BACKGROUND DATA.: MM is one of representative hematologic malignancies that cause skeletal-related events (SREs) and dysregulation of bone remodeling is known as a key pathomechanism of disease progression and skeletal-related events. However, decreased proliferation of MM at fracture sites is frequently noted in clinical situations regardless of systemic disease activity. METHODS Co-culture under various conditions was used to investigate effects of increased osteoblastic activity on survival and proliferation of MM plasma cells. MM plasma cells were cultured in culture media (control) and co-cultured with human mesenchymal stem cells (hMSCs, group I), osteoblasts (OBs) induced from hMSCs (group II) or bone morphogenic protein-2 (BMP-2, group III). Proliferation measured as extracellular signal-regulated kinase (ERK) and immunoglobulin G (Ig G) expression and apoptosis measured as fluorescence-activated cell sorting (FACS) with annexin V method, caspase-3, and stat-3 expression were assessed for cultured MM plasma cells, along with expression of sclerostin. RESULTS After 72 hours of co-culture, group II and III showed decreased ERK expression compared with controls. Lower Ig G expression was also noted for groups II and III compared with controls. Group I did not show significantly decreased Ig G and ERK expression compared with controls. Expressions of caspase-3 in groups II and III were higher than controls. Co-culture with hMSCs showed decreased caspase-3 expression compared with control. FACS with annexin V showed higher apoptosis in groups II and III. Sclerostin expression was also decreased in osteoblastic conditions compared with the control and hMSCs co-culture condition. CONCLUSION Collectively, our data suggest that increased osteoblastic conditions may provide not only prevention of SREs but also anti-tumor effects on MM cells in the bone marrow environment. LEVEL OF EVIDENCE N/A.
Collapse
|
5
|
Ding B, Zhang W, Wu X, Wang J, Xie C, Huang X, Zhan S, Zheng Y, Huang Y, Xu N, Ding X, Gao S. DR5 mAb-conjugated, DTIC-loaded immuno-nanoparticles effectively and specifically kill malignant melanoma cells in vivo. Oncotarget 2018; 7:57160-57170. [PMID: 27494835 PMCID: PMC5302980 DOI: 10.18632/oncotarget.11014] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2016] [Accepted: 07/19/2016] [Indexed: 12/18/2022] Open
Abstract
We combined chemo- and immunotherapies by constructing dual therapeutic function immuno-nanoparticles (NPs) consisting of death receptor 5 monoclonal antibody (DR5 mAb)-conjugated nanoparticles loaded with dacarbazine (DTIC) (DTIC-NPs-DR5 mAb). We determined the in vivo targeting specificity of DTIC-NPs-DR5 mAb by evaluating distribution in tumor-bearing nude mice using a real-time imaging system. Therapeutic efficacy was assessed in terms of its effect on tumor volume, survival time, histomorphology, microvessel density (MVD), and apoptotic index (AI). Systemic toxicity was evaluated by measuring white blood cells (WBC) counts, alanine aminotransferase (ALT) levels, and creatinine clearance (CR).In vivo and ex vivo imaging indicates that DR5 mAb modification enhanced the accumulation of NPs within the xenograft tumor. DTIC-NPs-DR5 mAb inhibited tumor growth more effectively than DTIC or DR5 mAb alone, indicating that combining DTIC and DR5 mAb through pharmaceutical engineering achieves a better therapeutic effect. Moreover, the toxicity of DTIC-NPs-DR5 mAb was much lower than that of DTIC, implying that DR5 mAb targeting reduces nonspecific uptake of DTIC into normal tissue and thus decreases toxic side effects. These results demonstrate that DTIC-NPs-DR5 mAb is a safe and effective nanoparticle formulation with the potential to improve the efficacy and specificity of melanoma treatment.
Collapse
Affiliation(s)
- Baoyue Ding
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, PR China.,Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, PR China.,Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, USA
| | - Wei Zhang
- Department of Pharmaceutics, Shanghai Pulmonary Hospital, Tongji University, Shanghai, PR China
| | - Xin Wu
- Department of Pharmaceutics, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, PR China
| | - Jeffrey Wang
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, USA
| | - Chen Xie
- Department of Pharmaceutical Sciences, College of Pharmacy, Western University of Health Sciences, Pomona, USA
| | - Xuan Huang
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, PR China
| | - Shuyu Zhan
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, PR China
| | - Yongxia Zheng
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, PR China
| | - Yueyan Huang
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, PR China
| | - Ningyin Xu
- Department of Pharmaceutics, Medical College of Jiaxing University, Jiaxing, PR China
| | - Xueying Ding
- Department of Pharmaceutics, Shanghai General Hospital, Shanghai Jiaotong University, Shanghai, PR China
| | - Shen Gao
- Department of Pharmaceutics, Changhai Hospital, Second Military Medical University, Shanghai, PR China
| |
Collapse
|
6
|
Hao Y, Zhao F, Luo Y, Zhang M, Li S. Inhibitory effect of oridonin on proliferation of RPMI8226 cells and the possible underlying mechanism. J TRADIT CHIN MED 2016; 36:225-30. [PMID: 27400478 DOI: 10.1016/s0254-6272(16)30031-0] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
OBJECTIVE To observe the effects of oridonin on proliferation and apoptosis of myeloma RPMI8226 cells and to investigate the potential underlying mechanisms. METHODS RPMI8226 cells were treated with various concentrations of oridonin. Cell proliferation was analyzed using the thiazolyl blue tetrazolium bromide method. Ultramicrostructure was observed by transmission electron microscopy. Annexin-V/PI staining and flow cytometry was performed to determine cell apoptosis. Expression of apoptosis-related proteins was evaluated by western blot analysis. RESULTS Oridonin suppressed the proliferation of RPMI8226 cells and induced apoptosis in a time-and dose-dependent manner. Transmission electron microscopy confirmed apoptotic morphology upon treatment with 20 μmol/L oridonin and western blot revealed decreased expressions of the apoptosis suppressors survivin, Bcl-2 and pro-caspase-3 proteins, and the increased expression of the apoptosis inducer Bax. CONCLUSION Our results show that oridonin exhibits an inhibitory effect on the proliferation of RPMI8226 cells and induces apoptosis. This is associated with altering the balance between Bcl-2 and Bax protein expressions and decreased survivin and pro-caspase-3 expressions.
Collapse
|
7
|
Lajmi N, Luetkens T, Yousef S, Templin J, Cao Y, Hildebrandt Y, Bartels K, Kröger N, Atanackovic D. Cancer-testis antigen MAGEC2 promotes proliferation and resistance to apoptosis in Multiple Myeloma. Br J Haematol 2015; 171:752-62. [DOI: 10.1111/bjh.13762] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/05/2015] [Accepted: 07/23/2015] [Indexed: 12/17/2022]
Affiliation(s)
- Nesrine Lajmi
- Hematology and Hematologic Malignancies; University of Utah; Huntsman Cancer Institute; Salt Lake City UT USA
- Oncology/Haematology/Bone Marrow Transplantation with the section Pneumology; University Medical Centre Hamburg-Eppendorf; Hamburg Germany
| | - Tim Luetkens
- Hematology and Hematologic Malignancies; University of Utah; Huntsman Cancer Institute; Salt Lake City UT USA
- Oncology/Haematology/Bone Marrow Transplantation with the section Pneumology; University Medical Centre Hamburg-Eppendorf; Hamburg Germany
| | - Sara Yousef
- Hematology and Hematologic Malignancies; University of Utah; Huntsman Cancer Institute; Salt Lake City UT USA
- Oncology/Haematology/Bone Marrow Transplantation with the section Pneumology; University Medical Centre Hamburg-Eppendorf; Hamburg Germany
- Stem Cell Transplantation; University Medical Centre Hamburg-Eppendorf; Hamburg Germany
| | - Julia Templin
- Hematology and Hematologic Malignancies; University of Utah; Huntsman Cancer Institute; Salt Lake City UT USA
- Oncology/Haematology/Bone Marrow Transplantation with the section Pneumology; University Medical Centre Hamburg-Eppendorf; Hamburg Germany
| | - Yanran Cao
- Oncology/Haematology/Bone Marrow Transplantation with the section Pneumology; University Medical Centre Hamburg-Eppendorf; Hamburg Germany
| | - York Hildebrandt
- Stem Cell Transplantation; University Medical Centre Hamburg-Eppendorf; Hamburg Germany
| | - Katrin Bartels
- Oncology/Haematology/Bone Marrow Transplantation with the section Pneumology; University Medical Centre Hamburg-Eppendorf; Hamburg Germany
| | - Nicolaus Kröger
- Stem Cell Transplantation; University Medical Centre Hamburg-Eppendorf; Hamburg Germany
| | - Djordje Atanackovic
- Hematology and Hematologic Malignancies; University of Utah; Huntsman Cancer Institute; Salt Lake City UT USA
- Oncology/Haematology/Bone Marrow Transplantation with the section Pneumology; University Medical Centre Hamburg-Eppendorf; Hamburg Germany
| |
Collapse
|
8
|
Hafez MM, Al-Harbi NO, Al-Hoshani AR, Al-Hosaini KA, Al Shrari SD, Al Rejaie SS, Sayed-Ahmed MM, Al-Shabanah OA. Hepato-protective effect of rutin via IL-6/STAT3 pathway in CCl4-induced hepatotoxicity in rats. Biol Res 2015; 48:30. [PMID: 26062544 PMCID: PMC4477598 DOI: 10.1186/s40659-015-0022-y] [Citation(s) in RCA: 64] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/07/2014] [Accepted: 06/03/2015] [Indexed: 01/20/2023] Open
Abstract
Background Carbon tetrachloride (CCl4) induces hepatotoxicity in animal models, including the increased blood flow and cytokine accumulation that are characteristic of tissue inflammation. The present study investigates the hepato-protective effect of rutin on CCl4-induced hepatotoxicity in rats. Results Forty male Wistar rats were divided into four groups. Group I (control group) received 1 mL/kg of dimethyl sulfoxide intragastrically and 3 mL/kg olive oil intraperitoneally twice a week for 4 weeks. Group II received 70 mg/kg rutin intragastrically. Groups III and IV received CCl4 (3 mL/kg, 30 % in olive oil) intraperitoneally twice a week for 4 weeks. Group IV received 70 mg/kg rutin intragastrically after 48 h of CCl4 treatment. Liver enzyme levels were determined in all studied groups. Expression of the following genes were monitored with real-time PCR: interleukin-6 (IL-6), dual-specificity protein kinase 5 (MEK5), Fas-associated death domain protein (FADD), epidermal growth factor (EGF), signal transducer and activator of transcription 3 (STAT3), Janus kinase (JAK), B-cell lymphoma 2 (Bcl2) and B-cell lymphoma-extra-large (Bcl-XL). The CCl4 groups showed significant increases in biochemical markers of hepatotoxicity and up-regulation of expression levels of IL-6, Bcl-XL, MEK5, FADD, EGF, STAT3 and JAK compared with the control group. However, CCl4 administration resulted in significant down-regulation of Bcl2 expression compared with the control group. Interestingly, rutin supplementation completely reversed the biochemical markers of hepatotoxicity and the gene expression alterations induced by CCl4. Conclusion CCl4 administration causes alteration in expression of IL-6/STAT3 pathway genes, resulting in hepatotoxicity. Rutin protects against CCl4-induced hepatotoxicity by reversing these expression changes.
Collapse
Affiliation(s)
- Mohamed M Hafez
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Kingdom of Saudi Arabia.
| | - Naif O Al-Harbi
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Kingdom of Saudi Arabia.
| | - Ali Rashed Al-Hoshani
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Kingdom of Saudi Arabia.
| | - Khaled A Al-Hosaini
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Kingdom of Saudi Arabia.
| | - Shakir D Al Shrari
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Kingdom of Saudi Arabia.
| | - Salim S Al Rejaie
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Kingdom of Saudi Arabia.
| | - Mohamed M Sayed-Ahmed
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Kingdom of Saudi Arabia.
| | - Othman A Al-Shabanah
- Department of Pharmacology and Toxicology, College of Pharmacy, King Saud University, P.O. Box 2457, Riyadh, 11451, Kingdom of Saudi Arabia.
| |
Collapse
|
9
|
Sano E, Tashiro S, Tsumoto K, Ueda T. Differential Effects of IFN-β on the Survival and Growth of Human Vascular Smooth Muscle and Endothelial Cells. Biores Open Access 2015; 4:1-15. [PMID: 26309778 PMCID: PMC4497630 DOI: 10.1089/biores.2014.0052] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022] Open
Abstract
It has been documented that interferon (IFN)-β is effective against the genesis of atherosclerosis or hyperplastic arterial disease in animal model. The main mechanism of the efficacy was antiproliferative action on the growth of vascular smooth muscle cells (SMC). To understand more about the mechanisms that are responsible for the efficacy, we examined minutely the effects of IFN-β on the apoptosis and growth of vascular SMC and endothelial cells (EC). IFN-β enhanced SMC apoptosis in serum starved medium. Conversely, EC apoptosis induced by serum and growth factor deprivation was inhibited by IFN-β. The induction of SMC apoptosis and anti-apoptotic effect on EC linked to the expression of pro-apoptotic bax mRNA and caspase-3 activities. Anti-apoptotic bcl-2 mRNA was also up-regulated in EC. IFN-β inhibited SMC growth in a dose dependent manner. However, the growth of EC was rather enhanced by a low dose of IFNs. The antiproliferative effect on SMC associated with the activation of p21 and increase of G0/G1 arrested cells. The growth stimulation on EC was considered to link with increase of S and G2/M phase cells. SMC produced IFN-β in response to various stimulants. However, IFN-β was not induced in EC. These suggested that endogenous IFN-β from SMC may act on EC and affect to EC functions. In this study, it was clarified that IFN-β enhances SMC apoptosis and inhibits the EC apoptosis, and stimulates the EC growth. These effects were considered to contribute to a cure against hyperplastic arterial diseases as the mechanisms in the efficacy of IFN-β.
Collapse
Affiliation(s)
- Emiko Sano
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo , Chiba, Japan
| | - Shinya Tashiro
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo , Chiba, Japan . ; Department of Medical Proteomics Laboratory, Institute of Medical Science, The University of Tokyo , Tokyo, Japan
| | - Kouhei Tsumoto
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo , Chiba, Japan . ; Department of Medical Proteomics Laboratory, Institute of Medical Science, The University of Tokyo , Tokyo, Japan . ; Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo , Tokyo, Japan
| | - Takuya Ueda
- Department of Medical Genome Sciences, Graduate School of Frontier Sciences, The University of Tokyo , Chiba, Japan
| |
Collapse
|
10
|
Zhou W, Chen H, Hong X, Niu X, Lu Q. Knockdown of DNA methyltransferase-1 inhibits proliferation and derepresses tumor suppressor genes in myeloma cells. Oncol Lett 2014; 8:2130-2134. [PMID: 25289094 PMCID: PMC4186563 DOI: 10.3892/ol.2014.2481] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2014] [Accepted: 07/23/2014] [Indexed: 12/15/2022] Open
Abstract
DNA methyltransferases (including DNMT1, DNMT3A and DNMT3B), catalyze the transfer of methyl groups from S-adenosyl-l-methionine to cytosine position 5; this methylation in promoter regions silences gene expression. In addition, DNMT1 plays a critical role in the maintenance of genomic DNA methylation during DNA replication. In the present study, silencing of DNMT1 with siRNA was performed in RPMI-8226 human multiple myeloma (MM) cells, and the impact on gene methylation status and proliferation of the cells was analyzed. Upon DNMT1 downregulation, proliferation decreased significantly compared with that in the control, non-transfected cells. The expression of B-cell lymphoma 2 and nuclear factor κB proteins was also significantly reduced. Furthermore, nested methylation-specific polymerase chain reaction revealed that methylation of the tumor suppressor genes, suppressor of cytokine signaling 1 and p16, was significantly reduced upon DNMT1 knockdown. Our results suggest that DNMT1 silencing may be a promising strategy to consider during development of novel MM treatment strategies.
Collapse
Affiliation(s)
- Wenwen Zhou
- Department of Hematology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361000, P.R. China
| | - Huying Chen
- Department of Hematology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361000, P.R. China
| | - Xiuli Hong
- Department of Hematology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361000, P.R. China
| | - Xiaoqing Niu
- Department of Hematology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361000, P.R. China
| | - Quanyi Lu
- Department of Hematology, Zhongshan Hospital, Xiamen University, Xiamen, Fujian 361000, P.R. China
| |
Collapse
|
11
|
Cancer therapeutics: Targeting the apoptotic pathway. Crit Rev Oncol Hematol 2014; 90:200-19. [DOI: 10.1016/j.critrevonc.2013.12.012] [Citation(s) in RCA: 115] [Impact Index Per Article: 10.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/18/2013] [Revised: 12/05/2013] [Accepted: 12/12/2013] [Indexed: 01/20/2023] Open
|
12
|
Zhang S, Suvannasankha A, Crean CD, White VL, Chen CS, Farag SS. The novel histone deacetylase inhibitor, AR-42, inhibits gp130/Stat3 pathway and induces apoptosis and cell cycle arrest in multiple myeloma cells. Int J Cancer 2010; 129:204-13. [PMID: 20824695 DOI: 10.1002/ijc.25660] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/10/2010] [Accepted: 08/20/2010] [Indexed: 11/09/2022]
Abstract
Multiple myeloma (MM) remains incurable with current therapy, indicating the need for continued development of novel therapeutic agents. We evaluated the activity of a novel phenylbutyrate-derived histone deacetylase inhibitor, AR-42, in primary human myeloma cells and cell lines. AR-42 was cytotoxic to MM cells at a mean LC(50) of 0.18 ± 0.06 μmol/l at 48 hr and induced apoptosis with cleavage of caspases 8, 9 and 3, with cell death largely prevented by caspase inhibition. AR-42 downregulated the expression of gp130 and inhibited activation of STAT3, with minimal effects on the PI3K/Akt and MAPK pathways, indicating a predominant effect on the gp130/STAT-3 pathway. AR-42 also inhibited interleukin (IL)-6-induced STAT3 activation, which could not be overcome by exogenous IL-6. AR-42 also downregulated the expression of STAT3-regulated targets, including Bcl-xL and cyclin D1. Overexpression of Bcl-xL by a lentivirus construct partly protected against cell death induced by AR-42. The cyclin dependent kinase inhibitors, p16 and p21, were also significantly induced by AR-42, which together with a decrease in cyclin D1, resulted in G(1) and G(2) cell cycle arrest. In conclusion, AR-42 has potent cytotoxicity against MM cells mainly through gp130/STAT-3 pathway. The results provide rationale for clinical investigation of AR-42 in MM.
Collapse
Affiliation(s)
- Shuhong Zhang
- Division of Hematology and Oncology, Department of Internal Medicine, Indiana University School of Medicine, Indianapolis, IN, USA
| | | | | | | | | | | |
Collapse
|
13
|
Fu J, Wang P, Zhang X, Ju S, Li J, Li B, Yu S, Zhang J, Zhang X. Myeloma cells inhibit osteogenic differentiation of mesenchymal stem cells and kill osteoblasts via TRAIL-induced apoptosis. Arch Med Sci 2010; 6:496-504. [PMID: 22371791 PMCID: PMC3284062 DOI: 10.5114/aoms.2010.14459] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/30/2009] [Revised: 09/05/2009] [Accepted: 09/15/2009] [Indexed: 12/21/2022] Open
Abstract
INTRODUCTION Myeloma bone disease (MBD) is the result of the increased activity of osteoclasts (OCs), which is not accompanied by a comparable increase of osteoblast (OB) function, thus leading to enhanced bone resorption. Osteoblasts can also regulate osteoclast activity through expression of cytokines, such as receptor activator of nuclear factor-κB ligand (RANKL), which activates osteoclast differentiation, and osteoprotegerin (OPG), which inhibits RANKL by acting as a decoy receptor. MATERIAL AND METHODS Based on a series of 21 patients with multiple myeloma (MM) and human osteoblast cell line HFOB1.19, we provide evidence that the bone marrow-derived mesenchymal stem cells (BMMSCs) of patients with MM exhibit normal phenotype, but showed reduced efficiency to differentiate into OBs as compared with normal controls. RESULTS In vitro assays showed that MM cells inhibited the potential of osteogenic differentiation of BMMSCs from healthy controls and rendered the OBs sensitive to TRAIL-induced apoptosis. There was no evidence of the formation of tartrate-resistant acid phosphatase positive OCs. The osteogenic differentiation of HFOB1.19 was also inhibited in the presence of RPMI 8266 or XG7 MM cells, as confirmed by von Kossa and ALP staining. Osteoblast s induced from BMMSCs supported survival and proliferation of MM cells, especially when the MM cells were cultured in medium containing rhTRAIL and dexamethasone. Multiple myeloma cells proliferated and grew well in the presence of residual OBs. CONCLUSIONS Besides OCs, our results demonstrated that OBs and MM cells were dependent upon each other and made a microenvironment suitable for MM cells.
Collapse
Affiliation(s)
- Jinxiang Fu
- Department of Haematology, No. 2 Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Panjun Wang
- Department of Haematology, No. 2 Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Xiaohui Zhang
- Department of Haematology, No. 2 Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Suguang Ju
- Institute of Biotechnology, Soochow University, China
| | - Jie Li
- Department of Haematology, Affiliated Children's Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Binzhou Li
- Department of Haematology, No. 2 Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Sun Yu
- Department of Haematology, No. 2 Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | - Jianhua Zhang
- Department of Haematology, No. 2 Affiliated Hospital of Soochow University, Suzhou, Jiangsu Province, China
| | | |
Collapse
|
14
|
Minarik J, Scudla V, Ordeltova M, Bacovsky J, Pika T, Langova K. Prognostic significance of apoptotic index in multiple myeloma patients treated by conventional therapy and novel agents, thalidomide and bortezomib. Eur J Haematol 2009; 83:528-34. [DOI: 10.1111/j.1600-0609.2009.01321.x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022]
|
15
|
Grcević D, Kusec R, Kovacić N, Lukić A, Lukić IK, Ivcević S, Nemet D, Seiwerth RS, Ostojić SK, Croucher PI, Marusić A. Bone morphogenetic proteins and receptors are over-expressed in bone-marrow cells of multiple myeloma patients and support myeloma cells by inducing ID genes. Leuk Res 2009; 34:742-51. [PMID: 19926132 DOI: 10.1016/j.leukres.2009.10.016] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/21/2009] [Revised: 09/30/2009] [Accepted: 10/20/2009] [Indexed: 12/30/2022]
Abstract
We assessed the expression pattern and clinical relevance of BMPs and related molecules in multiple myeloma (MM). MM bone-marrow samples (n=32) had increased BMP4, BMP6, ACVR1 and ACVR2A, and decreased NOG expression compared with controls (n=15), with BMP6 having the highest sensitivity/specificity. Within MM bone-marrow, the source of BMPs was mainly CD138(+) plasma-cell population, and BMP6 and ACVR1 expression correlated with plasma-cell percentage. Using myeloma cell lines NCI H929 and Thiel we showed that BMPs induced ID1, ID2 and IL6, and suppressed CDKN1A and BAX gene expression, and BAX protein expression. Finally, BMPs partially protected myeloma cells from bortezomib- and TRAIL-induced apoptosis. We concluded that BMPs may be involved in MM pathophysiology and serve as myeloma cell biomarkers.
Collapse
Affiliation(s)
- Danka Grcević
- Department of Physiology and Immunology, University of Zagreb, School of Medicine, Zagreb, Croatia.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Perez LE, Parquet N, Shain K, Nimmanapalli R, Alsina M, Anasetti C, Dalton W. Bone marrow stroma confers resistance to Apo2 ligand/TRAIL in multiple myeloma in part by regulating c-FLIP. THE JOURNAL OF IMMUNOLOGY 2008; 180:1545-55. [PMID: 18209050 DOI: 10.4049/jimmunol.180.3.1545] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Apo2 ligand (Apo2L)/TRAIL induces apoptosis of cancer cells that express the specific receptors while sparing normal cells. Because the tumor microenvironment protects myeloma from chemotherapy, we investigated whether hemopoietic stroma induces resistance to Apo2L/TRAIL apoptosis in this disease. Apo2L/TRAIL-induced death was diminished in myeloma cell lines (RPMI 8226, U266, and MM1s) directly adhered to a human immortalized HS5 stroma cell line but not adhered to fibronectin. In a Transwell assay, with myeloma in the upper well and HS5 cells in the lower well, Apo2L/TRAIL apoptosis was reduced when compared with cells exposed to medium in the lower well. Using HS5 and myeloma patients' stroma-conditioned medium, we determined that soluble factor(s) produced by stroma-myeloma interactions are responsible for a reversible Apo2/TRAIL apoptosis resistance. Soluble factor(s) attenuated procaspase-8, procaspase-3, and poly(ADP-ribose) polymerase cleavage and diminished mitochondrial membrane potential changes without affecting Bcl-2 family proteins and/or Apo2L/TRAIL receptors. Soluble factor(s) increased the baseline levels of the anti-apoptotic protein c-FLIP in all cell lines tested. Inhibition of c-FLIP by means of RNA interference increased Apo2/TRAIL sensitivity in RPMI 8226 cells. Unlike direct adhesion to fibronectin, soluble factor(s) have no impact on c-FLIP redistribution within cellular compartments. Cyclohexamide restored Apo2L/TRAIL sensitivity in association with down-regulation of c-FLIP, suggesting that c-FLIP synthesis, not intracellular traffic, is essential for soluble factor(s) to regulate c-FLIP. Additionally, IL-6 conferred resistance to Apo2L/TRAIL-mediated apoptosis in association with increased c-FLIP levels. In conclusion, the immune cytotoxic effect of Apo2L/TRAIL can be restored at least in part by c-FLIP pathway inhibitors.
Collapse
Affiliation(s)
- Lia Elena Perez
- Blood and Marrow Transplantation Program, H. Lee Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| | | | | | | | | | | | | |
Collapse
|
17
|
Zhang S, Suvannasankha A, Crean CD, White VL, Johnson A, Chen CS, Farag SS. OSU-03012, a Novel Celecoxib Derivative, Is Cytotoxic to Myeloma Cells and Acts through Multiple Mechanisms. Clin Cancer Res 2007; 13:4750-8. [PMID: 17699852 DOI: 10.1158/1078-0432.ccr-07-0136] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE OSU-03012 is a novel celecoxib derivative, without cyclooxygenase-2 inhibitory activity, capable of inducing apoptosis in various cancer cells types, and is being developed as an anticancer drug. We investigated the in vitro activity of OSU-03012 in multiple myeloma (MM) cells. EXPERIMENTAL DESIGN U266, ARH-77, IM-9, and RPMI-8226, and primary myeloma cells were exposed to OSU-03012 for 6, 24, or 72 h. Cytotoxicity, caspase activation, apoptosis, and effects on intracellular signaling pathways were assessed. RESULTS OSU-03012 was cytotoxic to MM cells with mean LC50 3.69 +/- 0.23 and 6.25 +/- 0.86 micromol/L and at 24 h for primary MM cells and cell lines, respectively. As a known PDK-1 inhibitor, OSU-03012 inhibited the PI3K/Akt pathway with downstream effects on BAD, GSK-3beta, FoxO1a, p70S6K, and MDM-2. However, transfection of MM cells with constitutively active Akt failed to protect against cell death, indicating activity against other pathways is important. Phospho (p)-signal transducers and activators of transcription 3 and p-MAP/ERK kinase 1/2 were down-regulated, suggesting that OSU-03012 also inhibited the Janus-activated kinase 2/signal transducer and activator of transcription 3 and mitogen-activated protein kinase pathways. Although expression of Bcl-2 proteins was unchanged, OSU-03012 also down-regulated survivin and X-linked inhibitor of apoptosis (XIAP), and also induced G2 cell cycle arrest with associated reductions in cyclins A and B. Finally, although OSU-03012 induced cleavage of caspases 3, 8 and 9, caspase inhibition did not prevent cell death. CONCLUSIONS We conclude that OSU-03012 has potent activity against MM cells and acts via different mechanisms in addition to phosphoinositide-3-kinase/Akt pathway inhibition. These studies provide rationale for the clinical investigation of OSU-03012 in MM.
Collapse
Affiliation(s)
- Shuhong Zhang
- Division of Hematology and Oncology, Department of Internal Medicine, Indiana University School of Medicine, Indianapolis, Indiana 46202, USA
| | | | | | | | | | | | | |
Collapse
|
18
|
Gómez-Benito M, Martinez-Lorenzo MJ, Anel A, Marzo I, Naval J. Membrane expression of DR4, DR5 and caspase-8 levels, but not Mcl-1, determine sensitivity of human myeloma cells to Apo2L/TRAIL. Exp Cell Res 2007; 313:2378-88. [PMID: 17462628 DOI: 10.1016/j.yexcr.2007.03.018] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2006] [Revised: 03/15/2007] [Accepted: 03/18/2007] [Indexed: 11/16/2022]
Abstract
The improved recombinant form of the death ligand Apo2L/TRAIL (Apo2L/TRAIL.0) is not cytotoxic for normal human cells and is a good candidate for the therapy of multiple myeloma (MM), a B-cell neoplasia that remains incurable. We have analyzed the molecular determinants of myeloma sensitivity to Apo2L/TRAIL.0 in a number of MM cell lines, the mechanisms of resistance and a possible way of overcoming it. Expression of one death receptor for Apo2L/TRAIL (DR4 or DR5) is sufficient to transduce death signals, though DR5 was more efficient when both receptors were present. Membrane expression of decoy receptors (DcR1, DcR2) and intracellular levels of c-FLIP(L), XIAP and Mcl-1 were not predictive of resistance to Apo2L/TRAIL. Inhibition of Mcl-1 degradation did not prevent Apo2L/TRAIL-induced apoptosis. In IM-9 cells, resistance was associated to a reduced caspase-8 expression. U266 cells, though expressing significant levels of DR4 and caspase-8, were nevertheless resistant to Apo2L/TRAIL. This resistance could be overcome by co-treatment with valproic acid (VPA), a histone deacetylase inhibitor. VPA caused the redistribution of DR4 to plasma membrane lipid rafts and restored DR4 signaling. Overexpression of Mcl-1 in U266 cells did not prevent Apo2L/TRAIL cytotoxicity in VPA-sensitized cells. These results, taken together, support the possible use of Apo2L/TRAIL.0 in the treatment of MM.
Collapse
Affiliation(s)
- Maria Gómez-Benito
- Departamento de Bioquímica y Biologia Molecular y Celular, Facultad de Ciencias, Universidad de Zaragoza, Zaragoza, Spain
| | | | | | | | | |
Collapse
|
19
|
Trudel S, Stewart AK, Li Z, Shu Y, Liang SB, Trieu Y, Reece D, Paterson J, Wang D, Wen XY. The Bcl-2 family protein inhibitor, ABT-737, has substantial antimyeloma activity and shows synergistic effect with dexamethasone and melphalan. Clin Cancer Res 2007; 13:621-9. [PMID: 17255285 DOI: 10.1158/1078-0432.ccr-06-1526] [Citation(s) in RCA: 102] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE The aim of this study is to investigate the antimyeloma activity of a novel Bcl-2 family inhibitor, ABT-737, in preclinical treatment of multiple myeloma. EXPERIMENTAL DESIGN The antimyeloma activity of ABT-737 was evaluated in cultured myeloma cell lines and patient myeloma samples, and in a xenograft mouse myeloma model. Drug combination therapy using ABT-737 with other commonly used myeloma drugs was also investigated. RESULTS MY5 and JJN3 cell lines exhibited the most sensitivity to ABT-737 with an EC(50) of 0.2 and 0.5 micromol/L, respectively, with increased cell apoptosis and elevated activated caspase-3. We identified two distinct groups of myeloma patient samples that were either sensitive or resistant to the drug. Four of 15 patient bone marrow samples (27%) were highly sensitive to ABT-737 at doses of 0.25 and 0.5 micromol/L, which eliminated 80% to 90% of myeloma cells as a result of cellular apoptosis 3 days after drug treatment. ABT-737 showed a synergistic effect when combined with dexamethasone or melphalan in inducing myeloma cell death. Furthermore, the dexamethasone-resistant MM1(Dex)R myeloma cell line was highly sensitive to 0.2 micromol/L ABT-737. As determined by colony assay, little or no detectable toxicity to patient hematologic progenitor cells was observed at 1 micromol/L ABT-737. ABT-737 dose dependently suppressed tumor growth in a xenograft MY5 mouse model. CONCLUSIONS These studies show substantial antimyeloma activity of ABT-737 as a single agent or in combination with dexamethasone or melphalan and suggest a rationale for future clinical trials.
Collapse
Affiliation(s)
- Suzanne Trudel
- Department of Medical Oncology and Hematology, Princess Margaret Hospital, University Health Network, University of Toronto, 620 University Avenue, Toronto, Ontario, Canada
| | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Agnelli L, Fabris S, Bicciato S, Basso D, Baldini L, Morabito F, Verdelli D, Todoerti K, Lambertenghi-Deliliers G, Lombardi L, Neri A. Upregulation of translational machinery and distinct genetic subgroups characterise hyperdiploidy in multiple myeloma. Br J Haematol 2007; 136:565-73. [PMID: 17367409 DOI: 10.1111/j.1365-2141.2006.06467.x] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Karyotypic instability, including numerical and structural chromosomal aberrations, represents a distinct feature of multiple myeloma (MM). About 40-50% of patients display hyperdiploidy, defined by recurrent trisomies of non-random chromosomes. To molecularly characterise hyperdiploid (H) and nonhyperdiploid (NH) MM, we analysed the gene expression profiles of 66 primary tumours, and used fluorescence in situ hybridisation to investigate the major chromosomal alterations. The differential expression of 225 genes mainly involved in protein biosynthesis, transcriptional machinery and oxidative phosphorylation distinguished the 28 H-MM from the 38 NH-MM cases. The 204 upregulated genes in H-MM mapped mainly to the chromosomes involved in hyperdiploidy, and the 29% upregulated genes in NH-MM mapped to 16q. The identified transcriptional fingerprint was robustly validated on a publicly available gene expression dataset of 64 MM cases; and the global expression modulation of regions on the chromosomes involved in hyperdiploidy was verified using a self-developed non-parametric statistical method. H-MM could be further divided into two distinct molecular and transcriptional entities, characterised by the presence of trisomy 11 and 1q-extracopies/chromosome 13 deletion respectively. These data reinforce the importance of combining molecular cytogenetics and gene expression profiling to define a genomic framework for the study of MM pathogenesis and clinical management.
Collapse
Affiliation(s)
- Luca Agnelli
- Centro di Genetica Molecolare ed Espressione Genica, Fondazione IRCCS Ospedale Maggiore Policlinico, Mangiagalli e Regina Elena, Milan, Italy
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Maillard S, Gauduchon J, Marsaud V, Gouilleux F, Connault E, Opolon P, Fattal E, Sola B, Renoir JM. Improved antitumoral properties of pure antiestrogen RU 58668-loaded liposomes in multiple myeloma. J Steroid Biochem Mol Biol 2006; 100:67-78. [PMID: 16753295 DOI: 10.1016/j.jsbmb.2006.03.008] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 02/07/2006] [Accepted: 03/23/2006] [Indexed: 10/24/2022]
Abstract
In most of multiple myeloma (MM) cells, the "pure" antiestrogen (AE) RU 58668 (RU) induced either a G1-arrest (LP-1, OPM-2, NCI-H929, U266 cells) or apoptosis (RPMI 8226 cells). In RPMI 8226 cells, RU activates a caspase-dependent cell death pathway leading to the release of cytochrome c, the decrease of the essential MM survival factor Mcl-1, the cleavage of Bid and the activation of caspases-3 and -8. Incorporation of RU in pegylated cholesterol-containing liposomes allowed a controlled RU release, improving its anti-proliferative and apoptotic effects in cells. In RPMI 8226 xenografts, i.v. injected RU-liposomes but not free RU, exhibited antitumor activity. In vivo, RU-liposomes triggered the mitochondrial death pathway, concomitantly with a down-regulation of Mcl-1 and Bid cleavage. The decrease of CD34 immunoreactivity indicated a reduction of angiogenesis. The decrease of VEGF secretion in vitro supported a direct effect of RU on angiogenesis. These pro-apoptotic and antiangiogenic effects were explained by a prolonged exposure to the drug and to the endocytosis capacity of liposomes which might increase RU uptake and bypass a membrane export of free RU. Thus, these combined enhanced activities of RU-liposomes support that such a delivery of an AE may constitute a strategy of benefit for MM treatment.
Collapse
|
22
|
Liu J, Huang R, Lin D, Wu X, Peng J, Lin Q, Pan X, Zhang M, Hou M, Chen F. Apoptotic effect of oridonin on NB4 cells and its mechanism. Leuk Lymphoma 2005; 46:593-7. [PMID: 16019488 DOI: 10.1080/10428190400019800] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
The anti-proliferation effects of oridonin on acute promyelocytic leukemia (APL) cells and its mechanisms were studied in vitro. NB4 cells as well as fresh leukemia cells obtained from APL patients in culture medium were treated with different concentrations of oridonin. Cell growth inhibition, apoptosis and related pathways were assessed by MTT assay as well as flow cytometry (FCM) and western blot analysis. The data revealed that oridonin (over 16 micromol/L) could inhibit the growth of NB4 cells by induction of apoptosis. Marked changes of cell apoptosis were observed very clearly by using electron microscopy and DNA fragmentation analysis after the cells exposed to oridonin for 48 h; Western blotting showed cleavage of the caspase-3 zymogen protein (32-kDa) with the appearance of its 20-kDa subunit as well as a cleaved 89-kDa fragment of 116-kDa PARP when apoptosis occurred. The expression of Bcl-2 was down-regulated remarkably accompanied by the disruption of the mitochondrial membrane potential (delta(psi)m). The anti-proliferative and apoptosis-inducing effects by oridonin in fresh APL cells were also found remarkably using Trypan Blue dye exclusion method and Wright's staining. We concluded that oridoning has significant anti-proliferative and apoptosis-inducing effects on NB4 cells by activation of caspase-3 and cleavage of PARP as well as by down regulation of Bcl-2 and disruption of the delta(psi)m. Furthermore, oridonin demonstrated apparent cell growth inhibition effects on fresh APL cells in vitro. The results indicated that oridonin may serve as a potential anti-leukemia reagent.
Collapse
MESH Headings
- Antineoplastic Agents/pharmacology
- Apoptosis/drug effects
- Blotting, Western
- Caspase 3
- Caspases/drug effects
- Caspases/metabolism
- Cell Line, Tumor
- Cell Proliferation/drug effects
- Diterpenes/pharmacology
- Diterpenes, Kaurane
- Dose-Response Relationship, Drug
- Drug Screening Assays, Antitumor
- Humans
- Leukemia, Promyelocytic, Acute/chemically induced
- Leukemia, Promyelocytic, Acute/drug therapy
- Leukemia, Promyelocytic, Acute/metabolism
- Molecular Structure
- Proto-Oncogene Proteins c-bcl-2/drug effects
- Proto-Oncogene Proteins c-bcl-2/metabolism
Collapse
Affiliation(s)
- Jiajun Liu
- Department of Hematology, The Third Affiliated Hospital of Sun Yat-sen University, Guangzhou, People's Republic of China.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Hu Y, Bebb G, Tan S, Ng R, Yan H, Sartor JR, Mayer LD, Bally MB. Antitumor efficacy of oblimersen Bcl-2 antisense oligonucleotide alone and in combination with vinorelbine in xenograft models of human non-small cell lung cancer. Clin Cancer Res 2005; 10:7662-70. [PMID: 15569999 DOI: 10.1158/1078-0432.ccr-04-1036] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Overexpression of Bcl-2 protein in cancer cells can inhibit programmed cell death and engender chemoresistance. Reducing Bcl-2 protein levels by using antisense oligonucleotides targeting the gene message can increase the sensitivity of cancer cells to cytotoxic agents. The objective of this work was to investigate the antitumor efficacy of the Bcl-2 antisense oligonucleotide oblimersen (Genasense; G3139), alone and in combination with vinorelbine (VNB), in an ectopic and orthotopic xenograft model of NCI-H460 human non-small-cell lung cancer. In addition to assessing therapeutic effect, Bcl-2 protein expression in tumor tissue isolated from lung and heart was measured. In the ectopic xenograft model, oblimersen at 5 and 10 mg/kg significantly inhibited tumor growth compared with saline-treated control groups, and furthermore, the antitumor effect of oblimersen was associated with down-regulation of Bcl-2 protein in isolated tumor tissue. Moreover, the combination of oblimersen with VNB was more active in inhibiting tumor growth than either drug used alone. In the orthotopic model, oblimersen treatment (5 mg/kg) increased the median survival time of mice to 33 days in comparison with a median survival time of 21 days in the control animals. With this model, the anticancer effect was demonstrated by assessing tumor growth in lung and heart tissues by hematoxylin and eosin staining and Bcl-2 expression by immunohistochemistry. When VNB at 5 mg/kg was combined with oblimersen administered at 5 mg/kg, 33% of mice survived more than 90 days. These data suggest that the combination of oblimersen and VNB may provide enhanced antitumor activities against non-small-cell lung cancer.
Collapse
Affiliation(s)
- YanPing Hu
- Genzyme Corporation, Framingham, Massachusetts, USA
| | | | | | | | | | | | | | | |
Collapse
|
24
|
Abstract
Multiple myeloma (MM) is a malignancy of terminally differentiated plasma cells. MM cells localize to the bone marrow, where cell adhesion-mediated autocrine or paracrine activation of various cytokines, such as interleukin 6, insulin-like growth factor 1, and interferon alpha, results in their accumulation mainly because of loss of critical apoptotic controls. Resistance to apoptosis, a genetically regulated cell death process, may play a critical role in both pathogenesis and resistance to treatment of MM. Abnormalities in regulation and execution of apoptosis can contribute to tumor initiation, progression, as well as to tumor resistance to various therapeutic agents. Apoptosis is executed via 2 main pathways that lead to activation of caspases: the death receptor (extrinsic) pathway and the mitochondrial (intrinsic) pathway. Ionizing radiation and chemotherapeutic agents act primarily through the intrinsic pathway, in which mitochondria play the central role. Various therapeutic modalities that are effective in MM modulate levels of the proapoptotic and antiapoptotic Bcl-2 family of proteins and of inhibitors of apoptosis, expression of which is primarily regulated by p53, nuclear factor KB, and STAT (signal transducers and activators of transcription) factors. This review focuses on the key concepts and some of the most recent studies of signaling pathways regulated in MM and summarizes what is known about the clinical role of these pathways.
Collapse
Affiliation(s)
- Marcela Oancea
- Department of Cancer Biology, Lerner Research Institute
- Department of Chemistry, Cleveland State University, Cleveland, Ohio, USA
| | | | | | - Alexandru Almasan
- Department of Cancer Biology, Lerner Research Institute
- Department of Radiation Oncology, Taussig Cancer Center, Cleveland Clinic Foundation
- Correspondence and reprint requests: Alex Almasan, PhD, Departments of Cancer Biology and Radiation Oncology, NB40, Cleveland Clinic Foundation, Cleveland, OH 44195, USA; 1-216-444-9970; fax: 1-216-445-6269 (e-mail:
)
| |
Collapse
|
25
|
Haddad JJ. On the antioxidant mechanisms of Bcl-2: a retrospective of NF-kappaB signaling and oxidative stress. Biochem Biophys Res Commun 2004; 322:355-63. [PMID: 15325238 DOI: 10.1016/j.bbrc.2004.07.138] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/05/2004] [Indexed: 11/22/2022]
Abstract
Antioxidant and prooxidant signaling pathways are emanating as major players in, and regulators of, cell death and apoptosis. Redox conception of the critical role of oxidative stress in determining cell fate is being established-a foundation that craves deeper than the basic understanding of physiochemical interactions to extend beyond that into the realms of deciphering the molecular codes implicated with apoptosis. The proto-oncogene Bcl-2 is no stranger being a major player and decoder in controlling apoptosis, ostensibly via the regulation of redox equilibrium and disequilibrium. One of those potential mechanisms exhibited by Bcl-2 is its ability to counteract the detrimental effects of cell damage caused by free radicals, thereby gaining its well-known property of being an antioxidant. But the question is: what are the molecular mechanisms involved with the antioxidant role of Bcl-2 in the face of cell damage and apoptosis? Currently, a stance is being upheld in that the Bcl-2 antioxidant efficacy should be weighed against its ability to manipulate transcriptional control, through the regulation of specific transcription factors. NF-kappaB is no doubt one of the best candidates when it comes to the arena of oxidative stress, inflammation, and apoptosis. Therein, current themes in the burgeoning antioxidant role of Bcl-2 are exposed within the context of transcriptional control of NF-kappaB, thereby holding potential avenues for alleviating therapeutic approaches in the regulation of apoptosis.
Collapse
Affiliation(s)
- John J Haddad
- Severinghaus-Radiometer Research Laboratories, University of California, San Francisco, CA, USA.
| |
Collapse
|