1
|
Liu YQ, Chen G, Wang KW, Yan XJ, Zhan CP, Yu GF. UCF-101 ameliorates traumatic brain injury by promoting microglia M2 polarization via AMPK/NF-κB pathways in LPS-induced BV2 cells. J Mol Histol 2024; 56:61. [PMID: 39739143 DOI: 10.1007/s10735-024-10336-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/27/2023] [Accepted: 12/10/2024] [Indexed: 01/02/2025]
Abstract
Traumatic brain injury (TBI) is a common neurosurgical emergency. As a macrophage in brain, microglia involves in secondary TBI injury. UCF-101, an Omi/HtrA2 inhibitor, protects against neurological disorders. This study aims to investigate the effects of UCF-101 in TBI and its mechanism. Mouse microglia cell BV2 cells were exposed to 1 µg/mL LPS to construct TBI in vitro models. Following CCK8 assay, cells were treated with LPS + UCF-101 (2, 5, 10 µM), LPS + Compound C (AMPK inhibitor, 20 µM), and LPS + UCF-101 + Compound C groups. With lactate dehydrogenase (LDH) content detection, ELISA and qRT-PCR assays were used to measure proinflammatory factors. Biomarkers of M1 (CD16/32 and iNOS) and M2 phenotypes (CD206), as well as AMPK/NF-κB pathway-related protein expression were assessed by flow cytometry, immunofluorescence, and Western blot methods. There was a decrease in M1 phenotype biomarkers and an increase in M2 phenotype biomarkers after UCF-101 treatment. UCF-101 exposure reduced TNF-α, LDH, IL-1β, IL-6, IL-8, p-NF-κB p65/NF-κB p65, and activated p-AMPK α (T172)/AMPK α (T172) expression. Importantly, further Compound C treatment counteracted these effects of UCF-101. In conclusion, UCF-101 ameliorates TBI by promoting microglia M2 polarization via AMPK/NF-κB pathways in LPS-induced BV2 cells, providing solid scientific foundation for clinical application of UCF-101 in TBI treatment.
Collapse
Affiliation(s)
- Yong-Qi Liu
- The Second Clinical Medical School, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Gao Chen
- School of Medicine, Quzhou College of Technology, Quzhou, Zhejiang, China
| | - Ke-Wei Wang
- The Second Clinical Medical School, Zhejiang Chinese Medical University, Hangzhou, Zhejiang, China
| | - Xin-Jiang Yan
- Department of Neurosurgery, Quzhou People's Hospital, No. 100, Minjiang Avenue, High-speed Rail, New City, Quzhou, Zhejiang, China
| | - Cheng-Peng Zhan
- Department of Neurosurgery, Quzhou People's Hospital, No. 100, Minjiang Avenue, High-speed Rail, New City, Quzhou, Zhejiang, China
| | - Guo-Feng Yu
- Department of Neurosurgery, Quzhou People's Hospital, No. 100, Minjiang Avenue, High-speed Rail, New City, Quzhou, Zhejiang, China.
| |
Collapse
|
2
|
Spina A, Guidarelli A, Buffi G, Fiorani M, Cantoni O. Unveiling the link between NADPH oxidase 2 activation and mitochondrial superoxide formation in leukemic cell killing induced by arsenic trioxide. Pharmacol Res 2024; 211:107554. [PMID: 39694125 DOI: 10.1016/j.phrs.2024.107554] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/29/2024] [Revised: 12/06/2024] [Accepted: 12/13/2024] [Indexed: 12/20/2024]
Abstract
This study focused on the interplay between NADPH oxidase 2 (NOX 2) activation and mitochondrial superoxide (mitoO2.-) formation induced by clinically relevant concentrations of arsenic trioxide (ATO; As2O3) in acute promyelocytic leukemia (APL) cells. Carefully controlled inhibitor studies and small interfering RNA mediated downregulation of p47phox (a component of the NOX 2 complex) expression demonstrated that, in an APL cell line, ATO promotes upstream NOX 2 activation critically connected with the formation of mitoO2.- and with the ensuing mitochondrial permeability transition (MPT)-dependent apoptosis. Instead, acute myeloid leukemia (AML) cell lines respond to ATO with low NOX 2 activation, resulting in a state that is non-permissive for mitoO2.- formation. Consistently, through rescue experiments, we demonstrate that pharmacological stimulation of NOX 2 overcomes resistance in these cells, thereby initiating the same cascade of downstream events observed in APL cells. As a final note, several lines of evidence, including measurement of glutathione, catalase and glutathione peroxidase levels, indicated that the antioxidant machinery was similar in APL and AML cells. The results regarding nuclear factor erythroid 2 p45-related factor 2-dependent antioxidant responses were instead of more complex interpretation as NB4 cells appeared particularly responsive to ATO. Our findings allow a novel interpretation of the interplay between NOX 2 activation and mitoO2.- formation induced by ATO, ultimately steering leukemic cells towards MPT-dependent apoptosis. These mechanistic insights provide a rationale for the disparate responses of APL and AML cells to ATO, offering potential avenues for the development of therapeutic intervention tailored to specific leukemia subtypes.
Collapse
Affiliation(s)
- Andrea Spina
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Andrea Guidarelli
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Gloria Buffi
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Mara Fiorani
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy
| | - Orazio Cantoni
- Department of Biomolecular Sciences, University of Urbino Carlo Bo, Urbino, Italy.
| |
Collapse
|
3
|
Shah MA, Hamid A, Faheem HI, Rasul A, Baokbah TAS, Haris M, Yousaf R, Saleem U, Iqbal S, Alves MS, Khan Z, Hussain G, Alsharfi I, Khan H, Jeandet P. Uncovering the Anticancer Potential of Polydatin: A Mechanistic Insight. Molecules 2022; 27:7175. [PMID: 36364001 PMCID: PMC9656535 DOI: 10.3390/molecules27217175] [Citation(s) in RCA: 7] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 10/17/2022] [Accepted: 10/17/2022] [Indexed: 11/05/2022] Open
Abstract
Polydatin or 3-O-β-d-resveratrol-glucopyranoside (PD), a stilbenoid component of Polygonum cuspicadum (Polygonaceae), has a variety of biological roles. In traditional Chinese medicine, P. cuspicadum extracts are used for the treatment of infections, inflammation, and cardiovascular disorders. Polydatin possesses a broad range of biological activities including antioxidant, anti-inflammatory, anticancer, and hepatoprotective, neuroprotective, and immunostimulatory effects. Currently, a major proportion of the population is victimized with cervical lung cancer, ovarian cancer and breast cancer. PD has been recognized as a potent anticancer agent. PD could effectively inhibit the migration and proliferation of ovarian cancer cells, as well as the expression of the PI3K protein. The malignancy of lung cancer cells was reduced after PD treatments via targeting caspase 3, arresting cancer cells at the S phase and inhibiting NLRP3 inflammasome by downregulation of the NF-κB pathway. This ceases cell cycle, inhibits VEGF, and counteracts ROS in breast cancer. It also prevents cervical cancer by regulating epithelial-to-mesenchymal transition (EMT), apoptosis, and the C-Myc gene. The objective of this review is thus to unveil the polydatin anticancer potential for the treatment of various tumors, as well as to examine the mechanisms of action of this compound.
Collapse
Affiliation(s)
| | - Ayesha Hamid
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Hafiza Ishmal Faheem
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Azhar Rasul
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Tourki A. S. Baokbah
- Department of Medical Emergency Services, College of Health Sciences-AlQunfudah, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Muhammad Haris
- Faculty of Pharmaceutical Sciences, Universiteit Gent, Ghent 9000, Belgium
| | - Rimsha Yousaf
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Uzma Saleem
- Faculty of Pharmaceutical Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Shabnoor Iqbal
- Department of Zoology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Maria Silvana Alves
- Laboratory of Cellular and Molecular Bioactivity, Department of Pharmaceutical Sciences, Faculty of Pharmacy, Federal University of Juiz de Fora, Juiz de Fora 36036-900, Brazil
| | - Zahid Khan
- Department of Pharmacognosy, Faculty of Pharmacy, Federal Urdu University of Arts, Science & Technology, Karachi 75300, Pakistan
| | - Ghulam Hussain
- Department of Physiology, Faculty of Life Sciences, Government College University, Faisalabad 38000, Pakistan
| | - Ifat Alsharfi
- Department of Biology, Jamoum University College, Umm Al-Qura University, Makkah 21955, Saudi Arabia
| | - Haroon Khan
- Department of Pharmacy, Abdul Wali Khan University Mardan, Mardan 23200, Pakistan
| | - Philippe Jeandet
- Research Unit Induced Resistance and Plant Bioprotection, University of Reims Champagne-Ardenne, USC INRAe 1488, 51100 Reims, France
| |
Collapse
|
4
|
Hu X, Cai J, Zhu J, Lang W, Zhong J, Zhong H, Chen F. Arsenic trioxide potentiates Gilteritinib-induced apoptosis in FLT3-ITD positive leukemic cells via IRE1a-JNK-mediated endoplasmic reticulum stress. Cancer Cell Int 2020; 20:250. [PMID: 32565734 PMCID: PMC7298957 DOI: 10.1186/s12935-020-01341-5] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/03/2020] [Accepted: 06/12/2020] [Indexed: 02/06/2023] Open
Abstract
Background Acute myeloid leukemia (AML) patients with FMS-like tyrosine kinase 3-internal tandem duplication (FLT3-ITD) have a high relapse rate and poor prognosis. This study aims to explore the underlying mechanism of combining Gilteritinib with ATO at low concentration in the treatment of FLT3-ITD positive leukemias. Methods We used both in vitro and in vivo studies to investigate the effects of combination of Gilteritinib with ATO at low concentration on FLT3-ITD positive leukemias, together with the underlying molecular mechanisms of these processes. Results Combination of Gilteritinib with ATO showed synergistic effects on inhibiting proliferation, increasing apoptosis and attenuating invasive ability in FLT3-ITD-mutated cells and reducing tumor growth in nude mice. Results of western blot indicated that Gilteritinib increased a 160KD form of FLT3 protein on the surface of cell membrane. Detection of endoplasmic reticulum stress marker protein revealed that IRE1a and its downstream signal phosphorylated JNK were suppressed in Gilteritinib-treated FLT3-ITD positive cells. The downregulation of IRE1a induced by Gilteritinib was reversed with addition of ATO. Knockdown of IRE1a diminished the combinatorial effects of Gilteritinib plus ATO treatment and combination of tunicamycin (an endoplasmic reticulum pathway activator) with Gilteritinib achieved the similar effect as treatment with Gilteritinib plus ATO. Conclusions Thus, ATO at low concentration potentiates Gilteritinib-induced apoptosis in FLT3-ITD positive leukemic cells via IRE1a-JNK signal pathway, targeting IRE1a to cooperate with Gilteritinib may serve as a new theoretical basis on FLT3-ITD mutant AML treatment.
Collapse
Affiliation(s)
- Xiaoli Hu
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127 China
| | - Jiayi Cai
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127 China
| | - Jianyi Zhu
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127 China
| | - Wenjing Lang
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127 China
| | - Jihua Zhong
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127 China
| | - Hua Zhong
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127 China
| | - Fangyuan Chen
- Department of Hematology, Ren Ji Hospital, School of Medicine, Shanghai Jiao Tong University, 160 Pujian Road, Shanghai, 200127 China
| |
Collapse
|
5
|
mTOR Regulation of Metabolism in Hematologic Malignancies. Cells 2020; 9:cells9020404. [PMID: 32053876 PMCID: PMC7072383 DOI: 10.3390/cells9020404] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/06/2019] [Revised: 02/02/2020] [Accepted: 02/07/2020] [Indexed: 02/06/2023] Open
Abstract
Neoplastic cells rewire their metabolism, acquiring a selective advantage over normal cells and a protection from therapeutic agents. The mammalian Target of Rapamycin (mTOR) is a serine/threonine kinase involved in a variety of cellular activities, including the control of metabolic processes. mTOR is hyperactivated in a large number of tumor types, and among them, in many hematologic malignancies. In this article, we summarized the evidence from the literature that describes a central role for mTOR in the acquisition of new metabolic phenotypes for different hematologic malignancies, in concert with other metabolic modulators (AMPK, HIF1α) and microenvironmental stimuli, and shows how these features can be targeted for therapeutic purposes.
Collapse
|
6
|
Haybar H, Shahrabi S, Rezaeeyan H, Jodat H, Saki N. Strategies to inhibit arsenic trioxide-induced cardiotoxicity in acute promyelocytic leukemia. J Cell Physiol 2019; 234:14500-14506. [PMID: 30770558 DOI: 10.1002/jcp.28292] [Citation(s) in RCA: 28] [Impact Index Per Article: 4.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Revised: 12/19/2018] [Accepted: 12/20/2018] [Indexed: 02/06/2023]
Abstract
OBJECTIVE Arsenic trioxide (ATO) is a drug commonly used for the treatment of acute promyelocytic leukemia (APL). Although ATO has been shown to cause significant improvement in patients, it is associated with serious side effects, which sometimes lead to the patient's death. In this review paper, we examine the reports of ATO-induced cardiotoxicity in APL patients and evaluate the strategies to reduce the incidence of such toxicity. METHODS The key search terms were "arsenic trioxide," "acute promyelocytic leukemia," "cardiotoxicity," "molecular pathway," and "biomarker." RESULTS Studies have indicated the involvement of several molecular pathways in ATO-induced cardiotoxicity. These pathways increase the production of reactive oxygen species by interfering with intracellular calcium homeostasis as well as impairing the transfer of calcium into endoplasmic reticulum and mitochondria. On the other hand, increasing or decreasing expressions of some microRNAs (miRs) have been shown to play a role in cardiotoxicity. CONCLUSION Finally, it can be stated that given the essential role of molecular pathways in cardiotoxicity and considering the fact these pathways impair the regulation of miRs expression, identification of molecular pathways involved in ATO-induced cardiotoxicity aimed at targeting miRs could be a new therapeutic strategy to prevent cardiotoxicity.
Collapse
Affiliation(s)
- Habib Haybar
- Atherosclerosis Research Center, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Saeid Shahrabi
- Department of Biochemistry and Hematology, Faculty of Medicine, Semnan University of Medical Sciences, Semnan, Iran
| | - Hadi Rezaeeyan
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Hosein Jodat
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| | - Najmaldin Saki
- Thalassemia and Hemoglobinopathy Research Center, Research Institute of Health, Ahvaz Jundishapur University of Medical Sciences, Ahvaz, Iran
| |
Collapse
|
7
|
Samad N, Jabeen S, Imran I, Zulfiqar I, Bilal K. Protective effect of gallic acid against arsenic-induced anxiety-/depression- like behaviors and memory impairment in male rats. Metab Brain Dis 2019; 34:1091-1102. [PMID: 31119507 DOI: 10.1007/s11011-019-00432-1] [Citation(s) in RCA: 65] [Impact Index Per Article: 10.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 08/13/2018] [Accepted: 05/13/2019] [Indexed: 12/16/2022]
Abstract
The purpose of the present study is to determine the effects of gallic acid (GA) on sodium arsenite (iAS)-induced behavior deficits and memory alteration in male rats. Thirty six animals were divided in to 6 groups (six animals in each) (i) saline+saline; (ii) saline+GA (50 mg/kg); (iii) saline+ GA (100 mg/kg) (iv) iAS + saline; (v) iAS + GA(50 mg/kg); (vi) iAS + GA (100 mg/kg). Animals were treated with iAS (2.5 mg/kg/ml); GA (50 and 100 mg/kg/ml) and saline (0.9%; 1 ml/kg) for 4 weeks. Repeated administration of iAS increases immobility time in forced swim test and decreases time spent in open arm (elevated plus maze) and light box (light dark activity box test) suggests depression like and anxiety-like symptoms respectively. On the other hand, animals treated with iAS + GA decreases immobility time and increases time spent in open arm and light box than saline+iAS treated animals suggests anxiolytic and antidepressant-like behavior of GA. Repeated administration of iAS also involves in memory impairment as observed in the Morris water maze test that is reversed by co-administration of GA, indicates that GA also involves in the enhancement of memory. Brain malondialdehyde (MDA) levels, antioxidant enzymes and acetylcholinesterase (AChE) activities also observed in the present study. Results show that iAS produces oxidative stress by increasing lipid peroxidation and decreasing antioxidant enzyme activity. Conversely co-administration of GA produces antioxidant effects by normalization of oxidative stress induced by iAS. Alteration in iAS induced AChE activity is also reversed by GA. It is suggested that GA via its antioxidant potential, has protective effects on iAS induced behavioral deficits and memory alteration. The findings have a strong implication on iAS induced neurological diseases, such as depression, anxiety, Alzheimer's disease and dementia etc.
Collapse
Affiliation(s)
- Noreen Samad
- Department of Biochemistry, Faculty of Science, Bahauddin Zakariya University, Multan, 60800, Pakistan.
| | - Sadia Jabeen
- Department of Biochemistry, Faculty of Science, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Imran Imran
- Department of Pharmacology, Faculty of Pharmacy, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Iqra Zulfiqar
- Department of Biochemistry, Faculty of Science, Bahauddin Zakariya University, Multan, 60800, Pakistan
| | - Kainat Bilal
- Department of Biochemistry, Faculty of Science, Bahauddin Zakariya University, Multan, 60800, Pakistan
| |
Collapse
|
8
|
Wang Q, Zou C, Wang L, Gao X, Wu J, Tan S, Wu G. Doxorubicin and adjudin co-loaded pH-sensitive nanoparticles for the treatment of drug-resistant cancer. Acta Biomater 2019; 94:469-481. [PMID: 31141733 DOI: 10.1016/j.actbio.2019.05.061] [Citation(s) in RCA: 36] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2019] [Revised: 05/13/2019] [Accepted: 05/23/2019] [Indexed: 02/08/2023]
Abstract
Multi-drug resistance (MDR) of tumor is a major cause of chemotherapy failure. In this study, a pH-sensitive graft copolymer, poly(β-amino ester)-g-β-cyclodextrin (PBAE-g-β-CD), was synthesized via Michael addition polymerization and was employed to co-deliver doxorubicin (DOX), a chemotherapy agent, and adjudin (ADD), a mitochondrial inhibitor, in the form of dual-drug co-loaded nanoparticles (NPs). Specifically, DOX was conjugated to 1-adamantaneacetic acid (Aa) to generate a prodrug that was subsequently encapsulated in the cavity of cyclodextrin via host-guest interactions. In addition, ADD was encapsulated by poly(β-aminoester) (PBAE). The introduction of the Aa-d-α-tocopheryl polyethylene glycolsuccinate (TPGS) conjugate enhanced the biocompatibility and serum stability of the resulting NPs. The NPs can realize precise ratiometric control of drugs being loaded, increase cellular uptake of the drugs, induce mitochondrial dysfunction and augment tumor treatment efficiency by inducing apoptosis. Western blot and polymerase chain reaction analyses showed that inhibition of P-glycoprotein and X-linked inhibitor of apoptosis protein expression may underlie inhibition of tumor resistance mediated by NPs. The MCF-7/ADR xenograft tumor model also revealed that in comparison with DOX, the NPs exhibited satisfactory performance in promoting apoptosis of tumor cells and achieved high therapeutic outcomes for MDR tumors. STATEMENT OF SIGNIFICANCE: Combination chemotherapy is an effective way to overcome MDR of tumor. However, one of the major obstacles for successful combination chemotherapy is the co-loading, co-delivery and controlled release of two different drugs, whose chemo-physical properties may be totally different. In this study, a pH-sensitive NP system was designed to realize the co-loading and precise ratiometric control of DOX and ADD, as well as the programmed drug release. That is, ADD release was triggered by low pH in endo/lysosome after endocytosis and then DOX was hydrolyzed to achieve a sustained release in tumor cells. Therefore, the NPs exhibited an effectively growth inhibition against MDR cells both in vitro and in vivo via the synergistic effect of ADD and DOX, which provided a promising strategy for treatment of MDR cancer.
Collapse
|
9
|
Zhang T, Zhu X, Wu H, Jiang K, Zhao G, Shaukat A, Deng G, Qiu C. Targeting the ROS/PI3K/AKT/HIF-1α/HK2 axis of breast cancer cells: Combined administration of Polydatin and 2-Deoxy-d-glucose. J Cell Mol Med 2019; 23:3711-3723. [PMID: 30920152 PMCID: PMC6484306 DOI: 10.1111/jcmm.14276] [Citation(s) in RCA: 100] [Impact Index Per Article: 16.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/12/2018] [Revised: 11/19/2018] [Accepted: 02/25/2019] [Indexed: 12/28/2022] Open
Abstract
It is well established that cancer cells depend upon aerobic glycolysis to provide the energy they need to survive and proliferate. However, anti-glycolytic agents have yielded few positive results in human patients, in part due to dose-limiting side effects. Here, we discovered the unexpected anti-cancer efficacy of Polydatin (PD) combined with 2-deoxy-D-glucose (2-DG), which is a compound that inhibits glycolysis. We demonstrated in two breast cell lines (MCF-7 and 4T1) that combination treatment with PD and 2-DG induced cell apoptosis and inhibited cell proliferation, migration and invasion. Furthermore, we determined the mechanism of PD in synergy with 2-DG, which decreased the intracellular reactive oxygen (ROS) levels and suppressed the PI3K/AKT pathway. In addition, the combined treatment inhibited the glycolytic phenotype through reducing the expression of HK2. HK2 deletion in breast cancer cells thus improved the anti-cancer activity of 2-DG. The combination treatment also resulted in significant tumour regression in the absence of significant morphologic changes in the heart, liver or kidney in vivo. In summary, our study demonstrates that PD synergised with 2-DG to enhance its anti-cancer efficacy by inhibiting the ROS/PI3K/AKT/HIF-1α/HK2 signalling axis, providing a potential anti-cancer strategy.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Huazhong Agricultural UniversityWuhanPeople's Republic of China
| | - Xinying Zhu
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Huazhong Agricultural UniversityWuhanPeople's Republic of China
| | - Haichong Wu
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Huazhong Agricultural UniversityWuhanPeople's Republic of China
| | - Kangfeng Jiang
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Huazhong Agricultural UniversityWuhanPeople's Republic of China
| | - Gan Zhao
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Huazhong Agricultural UniversityWuhanPeople's Republic of China
| | - Aftab Shaukat
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Huazhong Agricultural UniversityWuhanPeople's Republic of China
| | - Ganzhen Deng
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Huazhong Agricultural UniversityWuhanPeople's Republic of China
| | - Changwei Qiu
- Department of Clinical Veterinary MedicineCollege of Veterinary Medicine, Huazhong Agricultural UniversityWuhanPeople's Republic of China
| |
Collapse
|
10
|
MicroRNA Involvement in Allergic and Non-Allergic Mast Cell Activation. Int J Mol Sci 2019; 20:ijms20092145. [PMID: 31052286 PMCID: PMC6539777 DOI: 10.3390/ijms20092145] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2019] [Revised: 04/28/2019] [Accepted: 04/29/2019] [Indexed: 01/01/2023] Open
Abstract
Allergic inflammation is accompanied by the coordinated expression of numerous genes and proteins that initiate, sustain, and propagate immune responses and tissue remodeling. MicroRNAs (miRNAs) are a large class of small regulatory molecules that are able to control the translation of target mRNAs and consequently regulate various biological processes at the posttranscriptional level. MiRNA profiles have been identified in multiple allergic inflammatory diseases and in the tumor microenvironment. Mast cells have been found to co-localize within the above conditions. More specifically, in addition to being essential in initiating the allergic response, mast cells play a key role in both innate and adaptive immunity as well as in modulating tumor growth. This review summarizes the possible role of various miRNAs in the above-mentioned processes wherein mast cells have been found to be involved. Understanding the role of miRNAs in mast cell activation and function may serve as an important tool in developing diagnostic as well as therapeutic approaches in mast cell-dependent pathological conditions.
Collapse
|
11
|
Benajiba L, Alexe G, Su A, Raffoux E, Soulier J, Hemann MT, Hermine O, Itzykson R, Stegmaier K, Puissant A. Creatine kinase pathway inhibition alters GSK3 and WNT signaling in EVI1-positive AML. Leukemia 2018; 33:800-804. [PMID: 30390009 DOI: 10.1038/s41375-018-0291-x] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2018] [Revised: 08/24/2018] [Accepted: 09/18/2018] [Indexed: 11/09/2022]
Affiliation(s)
- Lina Benajiba
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,The Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, MA, USA.,INSERM U1163 and CNRS 8254, Imagine Institute, Université Paris Saclay, Paris, France
| | - Gabriela Alexe
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA, USA.,The Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, MA, USA.,Bioinformatics Graduate Program, Boston University, Boston, MA, USA
| | - Angela Su
- INSERM UMR 944, Institut Universitaire d'Hématologie, Hôpital St. Louis, Paris, France
| | - Emmanuel Raffoux
- Département d'Hématologie, Hôpital Saint-Louis, Assistance Publique - Hopitaux de Paris, Paris, France
| | - Jean Soulier
- Département d'Hématologie, Hôpital Saint-Louis, Assistance Publique - Hopitaux de Paris, Paris, France
| | - Michael T Hemann
- Koch Institute for Integrative Cancer Research at Massachusetts Institute of Technology, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Olivier Hermine
- INSERM U1163 and CNRS 8254, Imagine Institute, Université Paris Saclay, Paris, France
| | - Raphael Itzykson
- INSERM UMR 944, Institut Universitaire d'Hématologie, Hôpital St. Louis, Paris, France.,Département d'Hématologie, Hôpital Saint-Louis, Assistance Publique - Hopitaux de Paris, Paris, France
| | - Kimberly Stegmaier
- Department of Pediatric Oncology, Dana-Farber Cancer Institute and Boston Children's Hospital, Harvard Medical School, Boston, MA, USA. .,The Broad Institute of Harvard University and Massachusetts Institute of Technology, Cambridge, MA, USA.
| | - Alexandre Puissant
- INSERM UMR 944, Institut Universitaire d'Hématologie, Hôpital St. Louis, Paris, France.
| |
Collapse
|
12
|
Biological Aspects of mTOR in Leukemia. Int J Mol Sci 2018; 19:ijms19082396. [PMID: 30110936 PMCID: PMC6121663 DOI: 10.3390/ijms19082396] [Citation(s) in RCA: 21] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2018] [Revised: 08/07/2018] [Accepted: 08/10/2018] [Indexed: 02/07/2023] Open
Abstract
The mammalian target of rapamycin (mTOR) is a central processor of intra- and extracellular signals, regulating many fundamental cellular processes such as metabolism, growth, proliferation, and survival. Strong evidences have indicated that mTOR dysregulation is deeply implicated in leukemogenesis. This has led to growing interest in the development of modulators of its activity for leukemia treatment. This review intends to provide an outline of the principal biological and molecular functions of mTOR. We summarize the current understanding of how mTOR interacts with microRNAs, with components of cell metabolism, and with controllers of apoptotic machinery. Lastly, from a clinical/translational perspective, we recapitulate the therapeutic results in leukemia, obtained by using mTOR inhibitors as single agents and in combination with other compounds.
Collapse
|
13
|
Inhibition of IGF1R enhances 2-deoxyglucose in the treatment of non-small cell lung cancer. Lung Cancer 2018; 123:36-43. [PMID: 30089593 DOI: 10.1016/j.lungcan.2018.06.026] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/09/2018] [Revised: 06/21/2018] [Accepted: 06/22/2018] [Indexed: 12/16/2022]
Abstract
OBJECTIVE We previously postulated that 2-deoxyglucose (2-DG) activates multiple pro-survival pathways through IGF1R to negate its inhibitory effect on glycolysis. Here, we evaluated whether IGF1R inhibitor synergizes with 2-DG to impede the growth of non-small cell lung cancer (NSCLC). MATERIALS AND METHODS The activation of IGF1R signaling was assessed by the phosphorylation of IGF1R and its downstream target AKT using immunoblot. Drug dose response and combination index analyses were carried out according to the method of Chou and Talalay. Flow cytometry was used to evaluate cell cycle progression. Apoptosis was monitored by caspase-3/PARP cleavages or Annexin V staining. A subcutaneous xenograft model was used to assess this combination in vivo. RESULTS 2-DG induces the phosphorylation of IGF1R in its kinase domain, which can be abolished by the IGF1R inhibitor BMS-754807. Furthermore, the combination of 2-DG and BMS-754807 synergistically inhibited the survival of several non-small cell lung cancer (NSCLC) cell lines both in vitro and in vivo. The mechanistic basis of this synergy was cell line-dependent, and LKB1-inactivated EKVX cells underwent apoptosis following treatment with a subtoxic dose of 2-DG and BMS-754807. For these cells, the restoration of LKB1 kinase activity suppressed apoptosis induced by this combination but enhanced G1 arrest. In H460 cells, the addition of 2-DG did not enhance the low level of apoptosis induced by BMS-754807. However, treatment with 0.75 μM of BMS-754807 resulted in the accumulation of H460 cells with 8n-DNA content without affecting cell density increases. Hence, H460 cells may escape BMS-754807-induced G2/M cell cycle arrest through polyploidy. The inclusion of 2-DG blocked formation of the 8n-DNA cell population and restored G2/M phase cell cycle arrest. CONCLUSION The combination of 2-DG and IGF1R inhibitor BMS-754807 may be used to suppress the proliferation of NSCLC tumors through different mechanisms.
Collapse
|
14
|
Chen QY, Costa M. PI3K/Akt/mTOR Signaling Pathway and the Biphasic Effect of Arsenic in Carcinogenesis. Mol Pharmacol 2018; 94:784-792. [PMID: 29769245 PMCID: PMC5994485 DOI: 10.1124/mol.118.112268] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2018] [Accepted: 04/30/2018] [Indexed: 12/22/2022] Open
Abstract
Arsenic is a naturally occurring, ubiquitous metalloid found in the Earth’s crust. In its inorganic form, arsenic is highly toxic and carcinogenic and is widely found across the globe and throughout the environment. As an International Agency for Research on Cancer–defined class 1 human carcinogen, arsenic can cause multiple human cancers, including liver, lung, urinary bladder, skin, kidney, and prostate. Mechanisms of arsenic-induced carcinogenesis remain elusive, and this review focuses specifically on the role of the PI3K/AKT/mTOR pathway in promoting cancer development. In addition to exerting potent carcinogenic responses, arsenic is also known for its therapeutic effects against acute promyelocytic leukemia. Current literature suggests that arsenic can achieve both therapeutic as well as carcinogenic effects, and this review serves to examine the paradoxical effects of arsenic, specifically through the PI3K/AKT/mTOR pathway. Furthermore, a comprehensive review of current literature reveals an imperative need for future studies to establish and pinpoint the exact conditions for which arsenic can, and through what mechanisms it is able to, differentially regulate the PI3K/AKT/mTOR pathway to maximize the therapeutic and minimize the carcinogenic properties of arsenic.
Collapse
Affiliation(s)
- Qiao Yi Chen
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York
| | - Max Costa
- Department of Environmental Medicine, New York University School of Medicine, Tuxedo, New York
| |
Collapse
|
15
|
Glycolytic inhibitor 2-Deoxy-d-Glucose activates migration and invasion in glioblastoma cells through modulation of the miR-7-5p/TFF3 signaling pathway. Biochem Biophys Res Commun 2018; 499:829-835. [DOI: 10.1016/j.bbrc.2018.04.001] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2018] [Accepted: 04/01/2018] [Indexed: 02/06/2023]
|
16
|
Li D, Wei Y, Xu S, Niu Q, Zhang M, Li S, Jing M. A systematic review and meta-analysis of bidirectional effect of arsenic on ERK signaling pathway. Mol Med Rep 2018; 17:4422-4432. [PMID: 29328451 PMCID: PMC5802217 DOI: 10.3892/mmr.2018.8383] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2017] [Accepted: 11/21/2017] [Indexed: 01/05/2023] Open
Abstract
Arsenic is a toxic metal, which ultimately leads to cell apoptosis. ERK is considered a key transcriptional regulator of arsenic‑induced apoptosis. Due to a few controversial issues about arsenic‑mediated extracellular signal‑regulated MAP kinases (ERK) signaling, a meta‑analysis was performed. Subgroup analyses demonstrated that high doses (≥2 µmol/l) of arsenic increased the expression of Ras, ERK, ERK1, ERK2, phosphorylated (p)‑ERK, p‑ERK1, and p‑ERK2, while low doses (<2 µmol/l) decreased the expression of Ras, ERK1, p‑ERK, and p‑ERK2 when compared to control groups. Long term exposure (>24 h) to arsenic led to inhibition of expression of ERK1, p‑ERK1, and p‑ERK2, whereas short‑term exposure (≤24 h) triggered the expression of ERK1, ERK2, p‑ERK, p‑ERK1, and p‑ERK2. Furthermore, normal cells exposed to arsenic exhibited higher production levels of Ras and p‑ERK. Conversely, exposure of cancer cells to arsenic showed a lower level of production of Ras and p‑ERK as well as higher level of p‑ERK1 and p‑ERK2 as compared to control group. Short‑term exposure of normal cells to high doses of arsenic may promote ERK signaling pathway. In contrast, long‑term exposure of cancer cells to low doses of arsenic may inhibit ERK signaling pathway. This study may be helpful in providing a theoretical basis for the diverging result of arsenic adverse effects on one hand and therapeutic mechanisms on the other concerning arsenic‑induced apoptosis.
Collapse
Affiliation(s)
- Dongjie Li
- Department of Public Health, School of Medicine, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Yutao Wei
- Department of Cardiothoracic Surgery, First Affiliated Hospital, School of Medicine, Shihezi University, Shihezi, Xinjiang 832000, P.R. China
| | - Shangzhi Xu
- Department of Public Health, School of Medicine, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Qiang Niu
- Department of Public Health, School of Medicine, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Mei Zhang
- Department of Public Health, School of Medicine, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Shugang Li
- Department of Public Health, School of Medicine, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| | - Mingxia Jing
- Department of Public Health, School of Medicine, Shihezi University, Shihezi, Xinjiang 832002, P.R. China
| |
Collapse
|
17
|
Ranftler C, Meisslitzer-Ruppitsch C, Neumüller J, Ellinger A, Pavelka M. Golgi apparatus dis- and reorganizations studied with the aid of 2-deoxy-D-glucose and visualized by 3D-electron tomography. Histochem Cell Biol 2016; 147:415-438. [PMID: 27975144 PMCID: PMC5359389 DOI: 10.1007/s00418-016-1515-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/07/2016] [Indexed: 12/31/2022]
Abstract
We studied Golgi apparatus disorganizations and reorganizations in human HepG2 hepatoblastoma cells by using the nonmetabolizable glucose analogue 2-deoxy-d-glucose (2DG) and analyzing the changes in Golgi stack architectures by 3D-electron tomography. Golgi stacks remodel in response to 2DG-treatment and are replaced by tubulo-glomerular Golgi bodies, from which mini-Golgi stacks emerge again after removal of 2DG. The Golgi stack changes correlate with the measured ATP-values. Our findings indicate that the classic Golgi stack architecture is impeded, while cells are under the influence of 2DG at constantly low ATP-levels, but the Golgi apparatus is maintained in forms of the Golgi bodies and Golgi stacks can be rebuilt as soon as 2DG is removed. The 3D-electron microscopic results highlight connecting regions that interlink membrane compartments in all phases of Golgi stack reorganizations and show that the compact Golgi bodies mainly consist of continuous intertwined tubules. Connections and continuities point to possible new transport pathways that could substitute for other modes of traffic. The changing architectures visualized in this work reflect Golgi stack dynamics that may be essential for basic cell physiologic and pathologic processes and help to learn, how cells respond to conditions of stress.
Collapse
Affiliation(s)
- Carmen Ranftler
- Center for Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstraße 17, 1090, Vienna, Austria
| | | | - Josef Neumüller
- Center for Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstraße 17, 1090, Vienna, Austria
| | - Adolf Ellinger
- Center for Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstraße 17, 1090, Vienna, Austria
| | - Margit Pavelka
- Center for Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstraße 17, 1090, Vienna, Austria.
| |
Collapse
|
18
|
de Blas E, Estañ MC, Del Carmen Gómez de Frutos M, Ramos J, Del Carmen Boyano-Adánez M, Aller P. Selected polyphenols potentiate the apoptotic efficacy of glycolytic inhibitors in human acute myeloid leukemia cell lines. Regulation by protein kinase activities. Cancer Cell Int 2016; 16:70. [PMID: 27610044 PMCID: PMC5015235 DOI: 10.1186/s12935-016-0345-y] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2016] [Accepted: 08/30/2016] [Indexed: 12/19/2022] Open
Abstract
Background The glycolysis inhibitor 2-deoxy-d-glucose (2-DG) is a safe, potentially useful anti-tumour drug, but its efficacy is normally low when used alone. Recent studies indicated that 2-DG stimulates the PI3K/Akt and MEK/ERK defensive pathways, which limits the apoptotic efficacy in tumour cell lines. We hypothesized that co-treatment with selected polyphenols could improve 2-DG-provoked apoptosis by preventing defensive kinase activation. Methods Cell proliferation was measured by cell counting or the MTT assay. Cell cycle, apoptosis and necrosis were determined by propidium iodide staining and/or annexin V labeling followed by flow cytometry. Mitochondria pore transition and depolarization were determined by calcein-ATM or rhodamine 123 labeling followed flow cytometry. Intracellular reactive oxygen species and GSH were determined by dichlorodihydrofluorescein diacetate or monochlorobimane labeling followed by flow cytometry or fluorimetry. Expression and phosphorylation of protein kinases were analyzed by the Western blot. Results (i) 2-DG-provoked apoptosis was greatly potentiated by co-treatment with the sub-lethal concentrations of the flavonoid quercetin in human HL60 acute myeloblastic leukemia cells. Allowing for quantitative differences, apoptosis potentiation was also obtained using NB4 promyelocytic and THP-1 promonocytic cells, using curcumin or genistein instead of quercetin, and using lonidamine instead of 2-DG, but not when 2-DG was substituted by incubation in glucose-free medium. (ii) Quercetin and 2-DG rapidly elicited the opening of mitochondria pore transition, which preceded the trigger of apoptosis. (iii) Treatments did not affect GSH levels, and caused disparate effects on reactive oxygen species generation, which did not match the changes in lethality. (iv) 2-DG and lonidamine stimulated defensive Akt and ERK phosphorylation/activation, while glucose starvation was ineffective. Polyphenols prevented the stimulation of Akt phosphorylation, and in some cases also ERK phosphorylation. In addition, quercetin and 2-DG stimulated GSK-3α,β phosphorylation/inactivation, although with different isoform specificity. The use of pharmacologic inhibitors confirmed the importance of these kinase modifications for apoptosis. Conclusions The present in vitro observations suggest that co-treatment with low concentrations of selected polyphenols might represent a manner of improving the poor anti-tumour efficacy of some glycolytic inhibitors, and that apoptosis potentiation may be at least in part explained by the regulation of defensive protein kinase activities. Electronic supplementary material The online version of this article (doi:10.1186/s12935-016-0345-y) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Elena de Blas
- Centro de Investigaciones Biológicas (CIB), Consejo Superior de Investigaciones Científicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| | - María Cristina Estañ
- Centro de Investigaciones Biológicas (CIB), Consejo Superior de Investigaciones Científicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain ; Instituto de Investigaciones Biomédicas Alberto Sols, Consejo Superior de Investigaciones Científicas, Universidad Autónoma de Madrid, Madrid, Spain
| | - María Del Carmen Gómez de Frutos
- Centro de Investigaciones Biológicas (CIB), Consejo Superior de Investigaciones Científicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain ; Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| | - Javier Ramos
- Centro de Investigaciones Biológicas (CIB), Consejo Superior de Investigaciones Científicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain ; Escuela Técnica Superior de Ingenieros Agrónomos, Universidad Politécnica, Madrid, Spain
| | - María Del Carmen Boyano-Adánez
- Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá, Alcalá de Henares, Madrid, Spain
| | - Patricio Aller
- Centro de Investigaciones Biológicas (CIB), Consejo Superior de Investigaciones Científicas (CSIC), Ramiro de Maeztu 9, 28040 Madrid, Spain
| |
Collapse
|
19
|
Sulforaphane effects on postinfarction cardiac remodeling in rats: modulation of redox-sensitive prosurvival and proapoptotic proteins. J Nutr Biochem 2016; 34:106-17. [DOI: 10.1016/j.jnutbio.2016.05.004] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2016] [Revised: 04/26/2016] [Accepted: 05/11/2016] [Indexed: 12/24/2022]
|
20
|
Resveratrol augments ER stress and the cytotoxic effects of glycolytic inhibition in neuroblastoma by downregulating Akt in a mechanism independent of SIRT1. Exp Mol Med 2016; 48:e210. [PMID: 26891914 PMCID: PMC4892869 DOI: 10.1038/emm.2015.116] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/09/2015] [Revised: 09/07/2015] [Accepted: 09/22/2015] [Indexed: 12/15/2022] Open
Abstract
Cancer cells typically display increased rates of aerobic glycolysis that are correlated with tumor aggressiveness and a poor prognosis. Targeting the glycolytic pathway has emerged as an attractive therapeutic route mainly because it should spare normal cells. Here, we evaluate the effects of combining the inhibition of glycolysis with application of the polyphenolic compound resveratrol (RSV) in neuroblastoma (NB) cancer cell lines. Inhibiting glycolysis with 2-deoxy-D-glucose (2-DG) significantly reduced NB cell viability and was associated with increased endoplasmic reticulum (ER) stress and Akt activity. Administration of 2-DG increased the expression of the ER molecular chaperones GRP78 and GRP94, the prodeath protein C/EBP homology protein (CHOP) and the phosphorylation of Akt at S473, T450 and T308. Combined treatment with both RSV and 2-DG reduced GRP78, GRP94 and Akt phosphorylation but increased CHOP and NB cell death when compared with the administration of 2-DG alone. The selective inhibition of Akt activity also decreased 2-DG-induced GRP78 and GRP94 expression and increased CHOP expression, suggesting that Akt can modulate ER stress. Protein phosphatase 1α (PP1α) was activated by RSV, as indicated by a reduction in PP1α phosphorylation at T320. Pretreatment of cells with tautomycin, a selective PP1α inhibitor, prevented the RSV-mediated decrease in Akt phosphorylation, suggesting that RSV enhances 2-DG-induced cell death by activating PP1 and downregulating Akt. The RSV-mediated inhibition of Akt in the presence of 2-DG was not prevented by the selective inhibition of SIRT1, a known target of RSV, indicating that the effects of RSV on this pathway are independent of SIRT1. We propose that RSV inhibits Akt activity by increasing PP1α activity, thereby potentiating 2-DG-induced ER stress and NB cell death.
Collapse
|
21
|
Gupta P, Jagavelu K, Mishra DP. Inhibition of NADPH Oxidase-4 Potentiates 2-Deoxy-D-Glucose-Induced Suppression of Glycolysis, Migration, and Invasion in Glioblastoma Cells: Role of the Akt/HIF1α/HK-2 Signaling Axis. Antioxid Redox Signal 2015; 23:665-81. [PMID: 25891245 DOI: 10.1089/ars.2014.5973] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
AIMS 2-Deoxy-d-glucose (2-DG), a synthetic glycolytic inhibitor, is currently under clinical evaluation as a promising anticancer agent. However, 2-DG treatment in cancer cells activates prosurvival Akt signaling that might limit its clinical efficacy. The NADPH oxidase 4 (Nox-4)/reactive oxygen species/Akt signaling is known to regulate survival, proliferation, infiltration, and invasion in glioblastomas (GBMs). The enhanced motility, invasiveness, and therapy resistance in GBMs are attributed to metabolic adaptation through increased aerobic glycolysis. Therefore, we hypothesized that inhibition of the Nox-4 might enhance 2-DG-induced suppression of glycolysis, migration, and invasion in GBM cells. RESULTS We identified the natural naphthoquinone compound shikonin as a potent inhibitor of the Nox-4/Akt signaling pathway. The combined treatment of shikonin+2-DG suppressed the glycolytic phenotype, migration, and invasion through modulation of the Akt/HIF1α/hexokinase-2 signaling axis in GBM cells. The combination also exhibited enhanced antiproliferative and antiangiogenic effects in vivo. INNOVATION Our data for the first time demonstrate that inhibition of the Nox-4-associated prosurvival signaling pathway by shikonin enhances the antiproliferative and antiangiogenic potential of 2-DG in GBM cells. CONCLUSION In summary, the combined inhibition of Nox-4 and glycolysis may have therapeutic implications for the management of GBMs.
Collapse
Affiliation(s)
- Priyanka Gupta
- 1 Endocrinology Division, Cell Death Research Laboratory, CSIR-Central Drug Research Institute , Lucknow, India
| | - Kumaravelu Jagavelu
- 2 Pharmacology Division, CSIR-Central Drug Research Institute , Lucknow, India
| | - Durga Prasad Mishra
- 1 Endocrinology Division, Cell Death Research Laboratory, CSIR-Central Drug Research Institute , Lucknow, India
| |
Collapse
|
22
|
Wu YT, Wang BJ, Miao SW, Gao JJ. Picropodophyllin inhibits the growth of Ewing's sarcoma cells through the insulin‑like growth factor‑1 receptor/Akt signaling pathway. Mol Med Rep 2015; 12:7045-50. [PMID: 26323364 DOI: 10.3892/mmr.2015.4266] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2014] [Accepted: 07/29/2015] [Indexed: 11/06/2022] Open
Abstract
Ewing's sarcoma (ES) is the second most common type of pediatric bone tumor, and is associated with a poor prognosis. Picropodophyllin (PPP), a novel selective inhibitor of insulin‑like growth factor‑1 receptor (IGF‑1R), is able to strongly inhibit various types of cancers. However, the effect of IGF‑1R on ES remains unclear. Following treatment with various concentrations of PPP for various times, cell viability was determined using an MTT assay. In addition, cell proliferation and apoptosis was investigated separately by bromodeoxyuridine staining and flow cytometry, respectively. The PPP‑associated signaling pathway was also investigated. The results of the present study suggested that PPP inhibited cell proliferation and viability of A673 and SK‑ES‑1 human Ewing's sarcoma cells in a dose- and time‑dependent manner. In addition, cell apoptosis rates were increased following treatment with PPP. Further investigation of the underlying mechanism revealed that PPP inhibited Akt phosphorylation. Fumonisin B1, an Akt‑specific activator, reversed the inhibitory effects of PPP on cell growth. Furthermore, the results suggested that PPP decreased the expression levels of IGF‑1R, a common activator of Akt signaling. PPP inhibited the growth of human Ewing's sarcoma cells by targeting the IGF‑1R/Akt signaling pathway. Therefore, PPP may prove useful in the development of an effective strategy for the treatment of Ewing's sarcoma.
Collapse
Affiliation(s)
- Yong-Tao Wu
- Department of Pediatric Orthopedics, Hong Hui Hospital, Xi'an Jiaotong University College Medicine, Xi'an, Shaanxi 710054, P.R. China
| | - Bao-Jun Wang
- Department of Orthopedics, Shaanxi Province Yangling Demonstration Zone Hospital, Yangling, Shaanxi 712100, P.R. China
| | - Sheng-Wu Miao
- Department of Pediatric Orthopedics, Hong Hui Hospital, Xi'an Jiaotong University College Medicine, Xi'an, Shaanxi 710054, P.R. China
| | - Jian-Jun Gao
- Department of Orthopedics, Shaanxi Province Yangling Demonstration Zone Hospital, Yangling, Shaanxi 712100, P.R. China
| |
Collapse
|
23
|
Lee M, Yoon JH. Metabolic interplay between glycolysis and mitochondrial oxidation: The reverse Warburg effect and its therapeutic implication. World J Biol Chem 2015; 6:148-61. [PMID: 26322173 PMCID: PMC4549759 DOI: 10.4331/wjbc.v6.i3.148] [Citation(s) in RCA: 107] [Impact Index Per Article: 10.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/19/2015] [Revised: 05/26/2015] [Accepted: 07/21/2015] [Indexed: 02/05/2023] Open
Abstract
Aerobic glycolysis, i.e., the Warburg effect, may contribute to the aggressive phenotype of hepatocellular carcinoma. However, increasing evidence highlights the limitations of the Warburg effect, such as high mitochondrial respiration and low glycolysis rates in cancer cells. To explain such contradictory phenomena with regard to the Warburg effect, a metabolic interplay between glycolytic and oxidative cells was proposed, i.e., the "reverse Warburg effect". Aerobic glycolysis may also occur in the stromal compartment that surrounds the tumor; thus, the stromal cells feed the cancer cells with lactate and this interaction prevents the creation of an acidic condition in the tumor microenvironment. This concept provides great heterogeneity in tumors, which makes the disease difficult to cure using a single agent. Understanding metabolic flexibility by lactate shuttles offers new perspectives to develop treatments that target the hypoxic tumor microenvironment and overcome the limitations of glycolytic inhibitors.
Collapse
|
24
|
Chuang IC, Yang CM, Song TY, Yang NC, Hu ML. The anti-angiogenic action of 2-deoxyglucose involves attenuation of VEGFR2 signaling and MMP-2 expression in HUVECs. Life Sci 2015; 139:52-61. [PMID: 26285173 DOI: 10.1016/j.lfs.2015.08.002] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Revised: 07/29/2015] [Accepted: 08/01/2015] [Indexed: 02/05/2023]
Abstract
AIMS 2-Deoxyglucose (2-DG) is a glucose analogue and has been shown to inhibit angiogenesis in human umbilical vascular endothelial cells (HUVECs) through interference with N-linked glycosylation. However, the anti-angiogenic mechanisms of 2-DG are not fully elucidated. MAIN METHODS We first employed an ex vivo rat aortic ring model to substantiate the anti-angiogenic action of 2-DG and then used HUVECs to investigate the molecular mechanism underlying such an action. KEY FINDINGS Results reveal that 2-DG (0.05-1.0mM) significantly inhibited tube formation in both rat aortic rings and HUVECs. 2-DG (0.1-1.0mM) also significantly inhibited cell invasion and migration, as well as the activity and mRNA and protein expression of matrix metalloproteinase (MMP)-2 in HUVECs. In addition, 2-DG (1.0mM) significantly inhibited mRNA and protein expression of vascular endothelial growth receptor 2 (VEGFR2) in a time-dependent manner. 2-DG also significantly inhibited the phosphorylation of the focal adhesion kinase (FAK) and mitogen-activated protein kinase (p38), the downstream molecules of VEGFR2. The effects of 2-DG on tube formation, MMP-2 activity, and VEGFR2 protein expression in HUVECs were reversed by mannose, an N-linked glycosylation precursor. Mannose also reversed 2-DG-induced accumulation of VEGFR2 in the endoplasmic reticulum. SIGNIFICANCE This ex vivo and in vitro study demonstrates that 2-DG inhibits angiogenesis with an action involving attenuation of VEGFR2 signaling and MMP-2 expression, possibly resulting from interference with N-linked glycosylation of VEGFR2. Further studies are needed to show that 2-DG inhibits VEGF-mediated angiogenesis or that the actual status of N-glycosylation of VEGFR2 is affected by the treatment.
Collapse
Affiliation(s)
- I-Chen Chuang
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Chih-Min Yang
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan
| | - Tuzz-Ying Song
- Department of BioIndustry Technology, Dayeh University, Changhua, Taiwan
| | - Nae-Cherng Yang
- School of Nutrition, Chung Shan Medical University, Taichung, Taiwan; Department of Nutrition, Chung Shan Medical University Hospital, Taichung, Taiwan.
| | - Miao-Lin Hu
- Department of Food Science and Biotechnology, National Chung Hsing University, Taichung, Taiwan; Agricultural Biotechnology Center, National Chung Hsing University, Taichung, Taiwan.
| |
Collapse
|
25
|
Wang Q, Zhao DY, Xu H, Zhou H, Yang QY, Liu F, Zhou GP. Down-regulation of microRNA-223 promotes degranulation via the PI3K/Akt pathway by targeting IGF-1R in mast cells. PLoS One 2015; 10:e0123575. [PMID: 25875646 PMCID: PMC4395227 DOI: 10.1371/journal.pone.0123575] [Citation(s) in RCA: 27] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2014] [Accepted: 03/05/2015] [Indexed: 02/07/2023] Open
Abstract
Background Mast cells play a central role in allergic and inflammatory disorders by inducing degranulation and inflammatory mediator release. Recent reports have shown that miRNAs play an important role in inflammatory response regulation. Therefore, the role of miR-223 in mast cells was investigated. Methods The expression of miR-223 was quantified by quantitative real-time polymerase chain reaction (qRT-PCR) in immunoglobulin E (IgE)-mediated mast cells. After successful miR-223 inhibition by transfection, degranulation was detected in IgE-mediated mast cells. The phosphorylation of IκB-α and Akt were examined using western blotting. NF-κB was tested using electrophoretic mobility shift assay. PI3K-inhibitor (LY294002) was used to investigate whether the PI3K/Akt pathway was essential for mast cell activation. The TargetScan database and a luciferase reporter system were used to identify whether insulin-like growth factor 1 receptor (IGF-1R) is a direct target of miR-223. Results MiR-223 expression was up-regulated in IgE-mediated mast cells, whereas its down-regulation promoted mast cell degranulation. Levels of IκB-α and Akt phosphorylation as well as NF-κB were increased in miR-223 inhibitor cells. LY294002 could block the PI3K/Akt signaling pathway and rescue the promotion caused by suppressing miR-223 in mast cells. IGF-1R was identified as a direct target of miR-223. Conclusions These findings suggest that down-regulation of miR-223 promotes degranulation via the PI3K/Akt pathway by targeting IGF-1R in mast cells.
Collapse
Affiliation(s)
- Quan Wang
- Department of Respiratory Medicine, Nanjing Children’s Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - De-Yu Zhao
- Department of Respiratory Medicine, Nanjing Children’s Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Hong Xu
- Department of Respiratory Medicine, Nanjing Children’s Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Hui Zhou
- Department of Pediatrics, Nanjing Maternal and Child Health Hospital of Nanjing Medical University, Nanjing, China
| | - Qian-Yuan Yang
- Department of Respiratory Medicine, Nanjing Children’s Hospital Affiliated to Nanjing Medical University, Nanjing, China
| | - Feng Liu
- Department of Respiratory Medicine, Nanjing Children’s Hospital Affiliated to Nanjing Medical University, Nanjing, China
- * E-mail: (FL); (GPZ)
| | - Guo-Ping Zhou
- Department of Pediatrics, the Frist Affiliated Hospital of Nanjing Medical University, Nanjing, China
- * E-mail: (FL); (GPZ)
| |
Collapse
|
26
|
Lozano-Santos C, Amigo-Jiménez I, Nova-Gurumeta S, Pérez-Sanz N, García-Pardo A, García-Marco JA. Arsenic trioxide synergistically potentiates the cytotoxic effect of fludarabine in chronic lymphocytic leukemia cells by further inactivating the Akt and ERK signaling pathways. Biochem Biophys Res Commun 2015; 461:243-8. [PMID: 25869069 DOI: 10.1016/j.bbrc.2015.04.007] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/28/2015] [Accepted: 04/02/2015] [Indexed: 11/25/2022]
Abstract
CLL remains an incurable disease, making it crucial to continue searching for new therapies efficient in all CLL cases. We have studied the effect of combining arsenic trioxide (ATO) with fludarabine, a frontline drug in CLL. We have found a synergistic interaction between 1 μM ATO and 5 μM fludarabine that significantly enhanced the cytotoxic effect of the individual drugs. Importantly, ATO sensitized fludarabine-resistant cells to the action of this drug. The mechanism behind this effect included the downregulation of phospho-Akt, phospho-ERK, and the Mcl-1/Bim and Bcl-2/Bax ratios. The combination of ATO and fludarabine partially overcame the survival effect induced by co-culturing CLL cells with stromal cells. Therefore, low concentrations of ATO combined with fludarabine may be an efficient therapeutic strategy in CLL patients.
Collapse
Affiliation(s)
- Carol Lozano-Santos
- Molecular Cytogenetics Unit, Hematology Department, Hospital Universitario Puerta de Hierro-Majadahonda and IDIPHIM, Madrid, Spain
| | - Irene Amigo-Jiménez
- Cellular and Molecular Medicine Department, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain
| | - Sara Nova-Gurumeta
- Molecular Cytogenetics Unit, Hematology Department, Hospital Universitario Puerta de Hierro-Majadahonda and IDIPHIM, Madrid, Spain
| | - Nuria Pérez-Sanz
- Molecular Cytogenetics Unit, Hematology Department, Hospital Universitario Puerta de Hierro-Majadahonda and IDIPHIM, Madrid, Spain
| | - Angeles García-Pardo
- Cellular and Molecular Medicine Department, Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas (CSIC), Madrid, Spain.
| | - José A García-Marco
- Molecular Cytogenetics Unit, Hematology Department, Hospital Universitario Puerta de Hierro-Majadahonda and IDIPHIM, Madrid, Spain.
| |
Collapse
|
27
|
Apoptotic efficacy of etomoxir in human acute myeloid leukemia cells. Cooperation with arsenic trioxide and glycolytic inhibitors, and regulation by oxidative stress and protein kinase activities. PLoS One 2014; 9:e115250. [PMID: 25506699 PMCID: PMC4266683 DOI: 10.1371/journal.pone.0115250] [Citation(s) in RCA: 37] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/18/2014] [Accepted: 11/20/2014] [Indexed: 12/18/2022] Open
Abstract
Fatty acid synthesis and oxidation are frequently exacerbated in leukemia cells, and may therefore represent a target for therapeutic intervention. In this work we analyzed the apoptotic and chemo-sensitizing action of the fatty acid oxidation inhibitor etomoxir in human acute myeloid leukemia cells. Etomoxir caused negligible lethality at concentrations up to 100 µM, but efficaciously cooperated to cause apoptosis with the anti-leukemic agent arsenic trioxide (ATO, Trisenox), and with lower efficacy with other anti-tumour drugs (etoposide, cisplatin), in HL60 cells. Etomoxir-ATO cooperation was also observed in NB4 human acute promyelocytic cells, but not in normal (non-tumour) mitogen-stimulated human peripheral blood lymphocytes. Biochemical determinations in HL60 cells indicated that etomoxir (25–200 µM) dose-dependently inhibited mitochondrial respiration while slightly stimulating glycolysis, and only caused marginal alterations in total ATP content and adenine nucleotide pool distribution. In addition, etomoxir caused oxidative stress (increase in intracellular reactive oxygen species accumulation, decrease in reduced glutathione content), as well as pro-apoptotic LKB-1/AMPK pathway activation, all of which may in part explain the chemo-sensitizing capacity of the drug. Etomoxir also cooperated with glycolytic inhibitors (2-deoxy-D-glucose, lonidamine) to induce apoptosis in HL60 cells, but not in NB4 cells. The combined etomoxir plus 2-deoxy-D-glucose treatment did not increase oxidative stress, caused moderate decrease in net ATP content, increased the AMP/ATP ratio with concomitant drop in energy charge, and caused defensive Akt and ERK kinase activation. Apoptosis generation by etomoxir plus 2-deoxy-D-glucose was further increased by co-incubation with ATO, which is apparently explained by the capacity of ATO to attenuate Akt and ERK activation. In summary, co-treatment with etomoxir may represent an interesting strategy to increase the apoptotic efficacy of ATO and (with some limitations) 2-deoxy-D-glucose which, although clinically important anti-tumour agents, exhibit low efficacy in monotherapy.
Collapse
|
28
|
Ranftler C, Meisslitzer-Ruppitsch C, Stangl H, Röhrl C, Fruhwürth S, Neumüller J, Pavelka M, Ellinger A. 2-Deoxy-D-glucose treatment changes the Golgi apparatus architecture without blocking synthesis of complex lipids. Histochem Cell Biol 2014; 143:369-80. [PMID: 25422148 DOI: 10.1007/s00418-014-1297-8] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/09/2014] [Indexed: 11/29/2022]
Abstract
The classic Golgi apparatus organization, an arrangement of highly ordered cisternal stacks with tubular-vesicular membrane specializations on both sides, is the functional image of a continuous flow of contents and membranes with input, metabolization, and output in a dynamic steady state. In response to treatment with 2-deoxy-D-glucose (2-DG), which lowers the cellular ATP level by about 70% within minutes, this organization is rapidly replaced by tubular-glomerular membrane convolutes described as Golgi networks and bodies. 2-DG is a non-metabolizable glucose analogue and competitive inhibitor of glycolysis, which has become attractive in the context of therapeutic approaches for several kinds of tumors specifically targeting glycolysis in cancer. With the question of whether the functions of the Golgi apparatus in lipid synthesis would be influenced by the 2-DG-induced Golgi apparatus reorganization, we focused on lipid metabolism within the Golgi bodies. For this, we applied a fluorophore-labeled short-chain ceramide (BODIPY-Cer) in various combinations with 2-DG treatment to HepG2 cell cultures and followed uptake, enrichment and metabolization to higher ordered lipids. The cellular ATP status in each experiment was controlled with a bioluminescence assay, and the response of the Golgi apparatus was tracked by immunostaining of the trans-Golgi network protein TGN46. For electron microscopy, the fluorescent BODIPY-Cer signals were converted into electron-dense precipitates by photooxidation of diaminobenzidine (DAB); DAB precipitates labeled trans-Golgi areas in control cultures but also compartments at the periphery of the Golgi bodies formed in response to 2-DG treatment, thus indicating that concentration of ceramide takes place in spite of the Golgi apparatus reorganization. Lipid analyses by thin-layer chromatography (TLC) performed in parallel showed that BODIPY-Cer is not only concentrated in compartments of the 2-DG-induced Golgi bodies but is partly metabolized to BODIPY-sphingomyelin. Both, uptake and condensation of BODIPY-Cer and its conversion to complex lipids indicate that functions of the Golgi apparatus in the cellular lipid metabolism persist although the classic Golgi apparatus organization is abolished.
Collapse
Affiliation(s)
- Carmen Ranftler
- Department of Cell Biology and Ultrastructure Research, Center for Anatomy and Cell Biology, Medical University of Vienna, Schwarzspanierstr. 17, 1090, Vienna, Austria
| | | | | | | | | | | | | | | |
Collapse
|
29
|
González-Gironès DM, Moncunill-Massaguer C, Iglesias-Serret D, Cosialls AM, Pérez-Perarnau A, Palmeri CM, Rubio-Patiño C, Villunger A, Pons G, Gil J. AICAR induces Bax/Bak-dependent apoptosis through upregulation of the BH3-only proteins Bim and Noxa in mouse embryonic fibroblasts. Apoptosis 2014; 18:1008-16. [PMID: 23605481 DOI: 10.1007/s10495-013-0850-6] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022]
Abstract
5-Aminoimidazole-4-carboxamide (AICA) riboside (AICAR) is a nucleoside analogue that is phosphorylated to 5-amino-4-imidazolecarboxamide ribotide (ZMP), which acts as an AMP mimetic and activates AMP-activated protein kinase (AMPK). It has been recently described that AICAR triggers apoptosis in chronic lymphocytic leukemia (CLL) cells, and its mechanism of action is independent of AMPK as well as p53. AICAR-mediated upregulation of the BH3-only proteins BIM and NOXA correlates with apoptosis induction in CLL cells. Here we propose mouse embryonic fibroblasts (MEFs) as a useful model to analyze the mechanism of AICAR-induced apoptosis. ZMP formation was required for AICAR-induced apoptosis, though direct Ampk activation with A-769662 failed to induce apoptosis in MEFs. AICAR potently induced apoptosis in Ampkα1 (-/-) /α2 (-/-) MEFs, demonstrating an Ampk-independent mechanism of cell death activation. In addition, AICAR acts independently of p53, as MEFs lacking p53 also underwent apoptosis normally. Notably, MEFs lacking Bax and Bak were completely resistant to AICAR-induced apoptosis, confirming the involvement of the mitochondrial pathway in its mechanism of action. Apoptosis was preceded by ZMP-dependent but Ampk-independent modulation of the mRNA levels of different Bcl-2 family members, including Noxa, Bim and Bcl-2. Bim protein levels were accumulated upon AICAR treatment of MEFs, suggesting its role in the apoptotic process. Strikingly, MEFs lacking both Bim and Noxa displayed high resistance to AICAR. These findings support the notion that MEFs are a useful system to further dissect the mechanism of AICAR-induced apoptosis.
Collapse
Affiliation(s)
- Diana M González-Gironès
- Departament de Ciències Fisiològiques II, Institut d'Investigació Biomèdica de Bellvitge IDIBELL-Universitat de Barcelona, Campus de Bellvitge, Pavelló de Govern, 4ª planta, L'Hospitalet de Llobregat, 08907 Barcelona, Catalonia, Spain
| | | | | | | | | | | | | | | | | | | |
Collapse
|
30
|
Guilbert C, Annis MG, Dong Z, Siegel PM, Miller WH, Mann KK. Arsenic trioxide overcomes rapamycin-induced feedback activation of AKT and ERK signaling to enhance the anti-tumor effects in breast cancer. PLoS One 2013; 8:e85995. [PMID: 24392034 PMCID: PMC3877392 DOI: 10.1371/journal.pone.0085995] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2013] [Accepted: 12/03/2013] [Indexed: 11/18/2022] Open
Abstract
Inhibitors of the mammalian target of rapamycin (mTORi) have clinical activity; however, the benefits of mTOR inhibition by rapamycin and rapamycin-derivatives (rapalogs) may be limited by a feedback mechanism that results in AKT activation. Increased AKT activity resulting from mTOR inhibition can be a result of increased signaling via the mTOR complex, TORC2. Previously, we published that arsenic trioxide (ATO) inhibits AKT activity and in some cases, decreases AKT protein expression. Therefore, we propose that combining ATO and rapamycin may circumvent the AKT feedback loop and increase the anti-tumor effects. Using a panel of breast cancer cell lines, we find that ATO, at clinically-achievable doses, can enhance the inhibitory activity of the mTORi temsirolimus. In all cell lines, temsirolimus treatment resulted in AKT activation, which was decreased by concomitant ATO treatment only in those cell lines where ATO enhanced growth inhibition. Treatment with rapalog also results in activated ERK signaling, which is decreased with ATO co-treatment in all cell lines tested. We next tested the toxicity and efficacy of rapamycin plus ATO combination therapy in a MDA-MB-468 breast cancer xenograft model. The drug combination was well-tolerated, and rapamycin did not increase ATO-induced liver enzyme levels. In addition, combination of these drugs was significantly more effective at inhibiting tumor growth compared to individual drug treatments, which corresponded with diminished phospho-Akt and phospho-ERK levels when compared with rapamycin-treated tumors. Therefore, we propose that combining ATO and mTORi may overcome the feedback loop by decreasing activation of the MAPK and AKT signaling pathways.
Collapse
Affiliation(s)
- Cynthia Guilbert
- Lady Davis Institute for Medical Research, McGill University, Montreal, Canada
| | - Matthew G. Annis
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Zhifeng Dong
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Peter M. Siegel
- Rosalind and Morris Goodman Cancer Research Centre, McGill University, Montreal, Canada
| | - Wilson H. Miller
- Lady Davis Institute for Medical Research, McGill University, Montreal, Canada
- Department of Oncology, McGill University, Montreal, Canada
| | - Koren K. Mann
- Lady Davis Institute for Medical Research, McGill University, Montreal, Canada
- Department of Oncology, McGill University, Montreal, Canada
- * E-mail:
| |
Collapse
|
31
|
Liu X, Gao Y, Yao H, Zhou L, Pei J, Sun L, Wang J, Sun D. p38 and Extracellular Signal-Regulated Kinases Activations have Opposite Effects on Primary-Cultured Rat Cerebellar Granule Neurons Exposed to Sodium Arsenite. J Biochem Mol Toxicol 2013; 28:143-8. [DOI: 10.1002/jbt.21546] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2013] [Revised: 11/06/2013] [Accepted: 11/24/2013] [Indexed: 12/20/2022]
Affiliation(s)
- Xiaona Liu
- Center for Endemic Disease Control; Chinese Center for Disease Control and Prevention; Harbin Medical University; Key Lab of Etiology and Epidemiology; Education Bureau of Heilongjiang Province and Ministry of Health; Harbin 150081 People's Republic of China
| | - Yanhui Gao
- Center for Endemic Disease Control; Chinese Center for Disease Control and Prevention; Harbin Medical University; Key Lab of Etiology and Epidemiology; Education Bureau of Heilongjiang Province and Ministry of Health; Harbin 150081 People's Republic of China
| | - Hongju Yao
- Center for Endemic Disease Control; Chinese Center for Disease Control and Prevention; Harbin Medical University; Key Lab of Etiology and Epidemiology; Education Bureau of Heilongjiang Province and Ministry of Health; Harbin 150081 People's Republic of China
| | - Lingwang Zhou
- Center for Endemic Disease Control; Chinese Center for Disease Control and Prevention; Harbin Medical University; Key Lab of Etiology and Epidemiology; Education Bureau of Heilongjiang Province and Ministry of Health; Harbin 150081 People's Republic of China
| | - Junrui Pei
- Center for Endemic Disease Control; Chinese Center for Disease Control and Prevention; Harbin Medical University; Key Lab of Etiology and Epidemiology; Education Bureau of Heilongjiang Province and Ministry of Health; Harbin 150081 People's Republic of China
| | - Liyan Sun
- Center for Endemic Disease Control; Chinese Center for Disease Control and Prevention; Harbin Medical University; Key Lab of Etiology and Epidemiology; Education Bureau of Heilongjiang Province and Ministry of Health; Harbin 150081 People's Republic of China
| | - Jing Wang
- Center for Endemic Disease Control; Chinese Center for Disease Control and Prevention; Harbin Medical University; Key Lab of Etiology and Epidemiology; Education Bureau of Heilongjiang Province and Ministry of Health; Harbin 150081 People's Republic of China
| | - Dianjun Sun
- Center for Endemic Disease Control; Chinese Center for Disease Control and Prevention; Harbin Medical University; Key Lab of Etiology and Epidemiology; Education Bureau of Heilongjiang Province and Ministry of Health; Harbin 150081 People's Republic of China
| |
Collapse
|
32
|
Calviño E, Estañ MC, Sánchez-Martín C, Brea R, de Blas E, Boyano-Adánez MDC, Rial E, Aller P. Regulation of death induction and chemosensitizing action of 3-bromopyruvate in myeloid leukemia cells: energy depletion, oxidative stress, and protein kinase activity modulation. J Pharmacol Exp Ther 2013; 348:324-35. [PMID: 24307199 DOI: 10.1124/jpet.113.206714] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
3-Bromopyruvate (3-BrP) is an alkylating, energy-depleting drug that is of interest in antitumor therapies, although the mechanisms underlying its cytotoxicity are ill-defined. We show here that 3-BrP causes concentration-dependent cell death of HL60 and other human myeloid leukemia cells, inducing both apoptosis and necrosis at 20-30 μM and a pure necrotic response at 60 μM. Low concentrations of 3-BrP (10-20 μM) brought about a rapid inhibition of glycolysis, which at higher concentrations was followed by the inhibition of mitochondrial respiration. The combination of these effects causes concentration-dependent ATP depletion, although this cannot explain the lethality at intermediate 3-BrP concentrations (20-30 μM). The oxidative stress caused by exposure to 3-BrP was evident as a moderate overproduction of reactive oxygen species and a concentration-dependent depletion of glutathione, which was an important determinant of 3-BrP toxicity. In addition, 3-BrP caused glutathione-dependent stimulation of p38 mitogen-activated protein kinase (MAPK), mitogen-induced extracellular kinase (MEK)/extracellular signal-regulated kinase (ERK), and protein kinase B (Akt)/mammalian target of rapamycin/p70S6K phosphorylation or activation, as well as rapid LKB-1/AMP kinase (AMPK) activation, which was later followed by Akt-mediated inactivation. Experiments with pharmacological inhibitors revealed that p38 MAPK activation enhances 3-BrP toxicity, which is conversely restrained by ERK and Akt activity. Finally, 3-BrP was seen to cooperate with antitumor agents like arsenic trioxide and curcumin in causing cell death, a response apparently mediated by both the generation of oxidative stress induced by 3-BrP and the attenuation of Akt and ERK activation by curcumin. In summary, 3-BrP cytotoxicity is the result of several combined regulatory mechanisms that might represent important targets to improve therapeutic efficacy.
Collapse
Affiliation(s)
- Eva Calviño
- Centro de Investigaciones Biológicas, Consejo Superior de Investigaciones Científicas, Madrid, Spain (E.C., M.C.E., C.S.-M., R.B., E.B., E.R., P.A.); and Departamento de Biología de Sistemas, Unidad de Bioquímica y Biología Molecular, Facultad de Medicina y Ciencias de la Salud, Universidad de Alcalá, Alcalá de Henares, Spain (M.C.B.-A.)
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Yu YH, Zhang L, Wu DS, Zhang Z, Huang FF, Zhang J, Chen XP, Liang DS, Zeng H, Chen FP. MiR-223 regulates human embryonic stem cell differentiation by targeting the IGF-1R/Akt signaling pathway. PLoS One 2013; 8:e78769. [PMID: 24250812 PMCID: PMC3826748 DOI: 10.1371/journal.pone.0078769] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2013] [Accepted: 09/16/2013] [Indexed: 11/19/2022] Open
Abstract
Currently, there are difficulties associated with the culturing of pluripotent human embryonic stem cells (hESCs), and knowledge regarding their regulatory mechanisms is limited. MicroRNAs (miRNAs) regulate gene expression and have critical functions in stem cell self-renewal and differentiation. Moreover, fibroblast growth factor (FGF) and the insulin-like growth factor receptor (IGF-1R) are key activators of signaling in hESCs. Based on the identification of complementary binding sites in miR-223 and IGF-1R mRNA, it is proposed that miR-223 acts as a local regulator of IGF-1R. Therefore, levels of miR-223 were detected in differentiated versus undifferentiated hESCs. In addition, proliferation, apoptosis, and differentiation were assayed in these two hESC populations and were compared in the presence of exogenous miR-223 and miR-223 inhibitor. Inhibition of miR-223 was found to maintain the undifferentiated state of hESCs, while addition of miR-223 induced differentiation. Furthermore, these effects were found to be likely dependent on IGF-1R/Akt signaling.
Collapse
Affiliation(s)
- Yan-Hui Yu
- Department of Hematology, Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Li Zhang
- Department of Hematology, West China Hospital, Si Chuan University, Chengdu, Sichuan, China
| | - Deng-Shu Wu
- Department of Hematology, Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Zheng Zhang
- Department of Pharmacology, School of Pharmaceutical Sciences, Central South University, Changsha, Hunan, China
| | - Fang-Fang Huang
- Department of Hematology, Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Jian Zhang
- Department of Hematology, Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
| | - Xiao-Ping Chen
- Pharmacogenetics Research Institute, Institute of Clinical Pharmacology, Central South University, Changsha, Hunan, China
| | - De-Sheng Liang
- State Key Laboratory of Medical Genetics of China, Central South University, Changsha, Hunan, China
| | - Hui Zeng
- Department of Hematology, Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
- * E-mail: (HZ); (FPC)
| | - Fang-Ping Chen
- Department of Hematology, Xiang-Ya Hospital, Central South University, Changsha, Hunan, China
- * E-mail: (HZ); (FPC)
| |
Collapse
|
34
|
Lu TH, Tseng TJ, Su CC, Tang FC, Yen CC, Liu YY, Yang CY, Wu CC, Chen KL, Hung DZ, Chen YW. Arsenic induces reactive oxygen species-caused neuronal cell apoptosis through JNK/ERK-mediated mitochondria-dependent and GRP 78/CHOP-regulated pathways. Toxicol Lett 2013; 224:130-40. [PMID: 24157283 DOI: 10.1016/j.toxlet.2013.10.013] [Citation(s) in RCA: 132] [Impact Index Per Article: 11.0] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2013] [Revised: 10/09/2013] [Accepted: 10/11/2013] [Indexed: 10/26/2022]
Abstract
Arsenic (As), a well-known high toxic metal, is an important environmental and industrial contaminant, and it induces oxidative stress, which causes many adverse health effects and diseases in humans, particularly in inorganic As (iAs) more harmful than organic As. Recently, epidemiological studies have suggested a possible relationship between iAs exposure and neurodegenerative disease development. However, the toxicological effects and underlying mechanisms of iAs-induced neuronal cell injuries are mostly unknown. The present study demonstrated that iAs significantly decreased cell viability and induced apoptosis in Neuro-2a cells. iAs also increased oxidative stress damage (production of malondialdehyde (MDA) and ROS, and reduction of Nrf2 and thioredoxin protein expression) and induced several features of mitochondria-dependent apoptotic signals, including: mitochondrial dysfunction, the activations of PARP and caspase cascades, and the increase in caspase-3 activity. Pretreatment with the antioxidant N-acetylcysteine (NAC) effectively reversed these iAs-induced responses. iAs also increased the phosphorylation of JNK and ERK1/2, but did not that p38-MAPK, in treated Neuro-2a cells. NAC and the specific JNK inhibitor (SP600125) and ERK1/2 inhibitor (PD98059) abrogated iAs-induced cell cytotoxicity, caspase-3/-7 activity, and JNK and ERK1/2 activation. Additionally, exposure of Neuro-2a cells to iAs triggered endoplasmic reticulum (ER) stress identified through several key molecules (GRP 78, CHOP, XBP-1, and caspase-12), which was prevented by NAC. Transfection with GRP 78- and CHOP-specific si-RNA dramatically suppressed GRP 78 and CHOP expression, respectively, and attenuated the activations of caspase-12, -7, and -3 in iAs-exposed cells. Therefore, these results indicate that iAs induces ROS causing neuronal cell death via both JNK/ERK-mediated mitochondria-dependent and GRP 78/CHOP-triggered apoptosis pathways.
Collapse
Affiliation(s)
- Tien-Hui Lu
- Department of Physiology, and Graduate Institute of Basic Medical Science, College of Medicine, China Medical University, Taichung 404, Taiwan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
35
|
Lu Y, Yuan H, Deng S, Wei Q, Guo C, Yi J, Wu J, Li R, Wen L, He Z, Yuan L. Arsanilic acid causes apoptosis and oxidative stress in rat kidney epithelial cells (NRK-52e cells) by the activation of the caspase-9 and -3 signaling pathway. Drug Chem Toxicol 2013; 37:55-62. [DOI: 10.3109/01480545.2013.806532] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022]
|