1
|
Tatarata QZ, Wang Z, Konopleva M. BCL-2 inhibition in acute myeloid leukemia: resistance and combinations. Expert Rev Hematol 2024; 17:935-946. [PMID: 39552410 DOI: 10.1080/17474086.2024.2429604] [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: 06/19/2024] [Accepted: 11/11/2024] [Indexed: 11/19/2024]
Abstract
INTRODUCTION The introduction of venetoclax has revolutionized the treatment landscape of acute myeloid leukemia, offering new therapeutic opportunities. However, the clinical response to venetoclax varies significantly between patients, with many experiencing limited duration of response. AREAS COVERED Identified resistance mechanisms include both intrinsic and acquired resistance to VEN. The former is associated with cell lineage and differentiation state. The latter includes dependency on alternative BCL-2 family anti-apoptotic protein(s) mediated by genetic, epigenetic, or post-translational mechanisms, mitochondrial and metabolic involvement, as well as microenvironment. Understanding these mechanisms is crucial for optimizing venetoclax-based therapies and enhancing treatment outcomes for patients with acute myeloid leukemia. This review aims to elucidate the primary mechanisms underlying resistance to venetoclax and explore current therapeutic strategies to overcome this challenge. EXPERT OPINION In patients with venetoclax resistance, alternative options include targeted combination therapies tailored to individual cases based on cytogenetics and prior treatments. Many of these therapies require further clinical investigation to validate their safety and efficacy.
Collapse
Affiliation(s)
- Qi Zhang Tatarata
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
- The Department of Medicine, SUNY Downstate Health Sciences University, Brooklyn, NY, USA
| | - Zhe Wang
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
| | - Marina Konopleva
- The Department of Leukemia, The University of Texas MD, Anderson Cancer Center, Houston, TX, USA
- Department of Oncology, Montefiore Medical Center and Albert Einstein College of Medicine, Bronx, NY, USA
| |
Collapse
|
2
|
Joghataie P, Ardakani MB, Sabernia N, Salary A, Khorram S, Sohbatzadeh T, Goodarzi V, Amiri BS. The Role of Circular RNA in the Pathogenesis of Chemotherapy-Induced Cardiotoxicity in Cancer Patients: Focus on the Pathogenesis and Future Perspective. Cardiovasc Toxicol 2024; 24:1151-1167. [PMID: 39158829 DOI: 10.1007/s12012-024-09914-w] [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: 09/21/2023] [Accepted: 08/11/2024] [Indexed: 08/20/2024]
Abstract
Cardiotoxicity is a serious challenge cancer patients face today. Various factors are involved in cardiotoxicity. Circular RNAs (circRNAs) are one of the effective factors in the occurrence and prevention of cardiotoxicity. circRNAs can lead to increased proliferation, apoptosis, and regeneration of cardiomyocytes by regulating the molecular pathways, as well as increasing or decreasing gene expression; some circRNAs have a dual role in cardiomyocyte regeneration or death. Identifying each of the pathways related to these processes can be effective on managing patients and preventing cardiotoxicity. In this study, an overview of the molecular pathways involved in cardiotoxicity by circRNAs and their effects on the downstream factors have been discussed.
Collapse
Affiliation(s)
- Pegah Joghataie
- Department of Cardiology, School of Medicine, Hazrat-E Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran
| | | | - Neda Sabernia
- Department of Internal Medicine, School of Medicine, Iran University of Medical Sciences, Tehran, Iran
| | | | | | - Tooba Sohbatzadeh
- Student Research Committee, School of Medicine, Alborz University of Medical Sciences, Alborz, Iran
| | - Vahid Goodarzi
- Department of Anesthesiology, Rasoul-Akram Medical Center, Iran University of Medical Sciences (IUMS), Tehran, Iran
| | - Bahareh Shateri Amiri
- Assistant Professor of Internal Medicine, Department of Internal Medicine, School of Medicine, Hazrat-E Rasool General Hospital, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
3
|
Arimura K, Kammer M, Rahman SMJ, Sheau-Chiann C, Zhao S, Heidi C, Eisenberg R, Zou Y, Antic S, Richmond B, Tagaya E, Grogan E, Massion P, Maldonado F. Elucidating the role of EPPK1 in lung adenocarcinoma development. BMC Cancer 2024; 24:441. [PMID: 38594604 PMCID: PMC11005125 DOI: 10.1186/s12885-024-12185-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/27/2023] [Accepted: 03/26/2024] [Indexed: 04/11/2024] Open
Abstract
BACKGROUND We recently found that epiplakin 1 (EPPK1) alterations were present in 12% of lung adenocarcinoma (LUAD) cases and were associated with a poor prognosis in early-stage LUAD when combined with other molecular alterations. This study aimed to identify a probable crucial role for EPPK1 in cancer development. METHODS EPPK1 mRNA and protein expression was analyzed with clinical variables. Normal bronchial epithelial cell lines were exposed to cigarette smoke for 16 weeks to determine whether EPPK1 protein expression was altered after exposure. Further, we used CRISPR-Cas9 to knock out (KO) EPPK1 in LUAD cell lines and observed how the cancer cells were altered functionally and genetically. RESULTS EPPK1 protein expression was associated with smoking and poor prognosis in early-stage LUAD. Moreover, a consequential mesenchymal-to-epithelial transition was observed, subsequently resulting in diminished cell proliferation and invasion after EPPK1 KO. RNA sequencing revealed that EPPK1 KO induced downregulation of 11 oncogenes, 75 anti-apoptosis, and 22 angiogenesis genes while upregulating 8 tumor suppressors and 12 anti-cell growth genes. We also observed the downregulation of MYC and upregulation of p53 expression at both protein and RNA levels following EPPK1 KO. Gene ontology enrichment analysis of molecular functions highlighted the correlation of EPPK1 with the regulation of mesenchymal cell proliferation, mesenchymal differentiation, angiogenesis, and cell growth after EPPK1 KO. CONCLUSIONS Our data suggest that EPPK1 is linked to smoking, epithelial to mesenchymal transition, and the regulation of cancer progression, indicating its potential as a therapeutic target for LUAD.
Collapse
Affiliation(s)
- Ken Arimura
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
- Department of Respiratory Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Michael Kammer
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - S M Jamshedur Rahman
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chen Sheau-Chiann
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Shilin Zhao
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Chen Heidi
- Department of Biostatistics, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Rosana Eisenberg
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Yong Zou
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Sanja Antic
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Bradley Richmond
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Etsuko Tagaya
- Department of Respiratory Medicine, Tokyo Women's Medical University, Tokyo, Japan
| | - Eric Grogan
- Department of Thoracic Surgery, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Pierre Massion
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA
| | - Fabien Maldonado
- Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University Medical Center, Nashville, TN, USA.
| |
Collapse
|
4
|
Stanland LJ, Ang HX, Hoj JP, Chu Y, Tan P, Wood KC, Luftig MA. CBF-Beta Mitigates PI3K-Alpha-Specific Inhibitor Killing through PIM1 in PIK3CA-Mutant Gastric Cancer. Mol Cancer Res 2023; 21:1148-1162. [PMID: 37493631 PMCID: PMC10811747 DOI: 10.1158/1541-7786.mcr-23-0034] [Citation(s) in RCA: 5] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2023] [Revised: 05/03/2023] [Accepted: 07/05/2023] [Indexed: 07/27/2023]
Abstract
PIK3CA is the second most mutated gene in cancer leading to aberrant PI3K/AKT/mTOR signaling and increased translation, proliferation, and survival. Some 4%-25% of gastric cancers display activating PIK3CA mutations, including 80% of Epstein-Barr virus-associated GCs. Small molecules, including pan-PI3K and dual PI3K/mTOR inhibitors, have shown moderate success clinically, due to broad on-target/off-tissue effects. Thus, isoform-specific and mutant selective inhibitors have been of significant interest. However, drug resistance is a problem and has affected success of new drugs. There has been a concerted effort to define mechanisms of resistance and identify potent combinations in many tumor types, though gastric cancer is comparatively understudied. In this study, we identified modulators of the response to the PI3Kα-specific inhibitor, BYL719, in PIK3CA-mutant GCs. We found that loss of NEDD9 or inhibition of BCL-XL conferred hypersensitivity to BYL719, through increased cell-cycle arrest and cell death, respectively. In addition, we discovered that loss of CBFB conferred resistance to BYL719. CBFB loss led to upregulation of the protein kinase PIM1, which can phosphorylate and activate several overlapping downstream substrates as AKT thereby maintaining pathway activity in the presence of PI3Kα inhibition. The addition of a pan-PIM inhibitor re-sensitized resistant cells to BYL719. Our data provide clear mechanistic insights into PI3Kα inhibitor response in PIK3CA-mutant gastric tumors and can inform future work as mutant-selective inhibitors are in development for diverse tumor types. IMPLICATIONS Loss of either NEDD9 or BCL-XL confers hypersensitivity to PI3K-alpha inhibition whereas loss of CBFB confers resistance through a CBFB/PIM1 signaling axis.
Collapse
Affiliation(s)
- Lyla J. Stanland
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine; Durham, NC, USA
| | - Hazel X. Ang
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine; Durham, NC, USA
| | - Jacob P. Hoj
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine; Durham, NC, USA
| | | | - Patrick Tan
- Duke-NUS Medical School Singapore; Singapore
- Genome Institute of Singapore, Agency for Science, Technology and Research; Singapore
| | - Kris C. Wood
- Department of Pharmacology and Cancer Biology, Duke University School of Medicine; Durham, NC, USA
| | - Micah A. Luftig
- Department of Molecular Genetics and Microbiology, Duke University School of Medicine; Durham, NC, USA
| |
Collapse
|
5
|
Wang W, Sun Y, Liu X, Kumar SK, Jin F, Dai Y. Dual-Targeted Therapy Circumvents Non-Genetic Drug Resistance to Targeted Therapy. Front Oncol 2022; 12:859455. [PMID: 35574302 PMCID: PMC9093074 DOI: 10.3389/fonc.2022.859455] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2022] [Accepted: 03/14/2022] [Indexed: 02/05/2023] Open
Abstract
The introduction of various targeted agents into the armamentarium of cancer treatment has revolutionized the standard care of patients with cancer. However, like conventional chemotherapy, drug resistance, either preexisting (primary or intrinsic resistance) or developed following treatment (secondary or acquired resistance), remains the Achilles heel of all targeted agents with no exception, via either genetic or non-genetic mechanisms. In the latter, emerging evidence supports the notion that intracellular signaling pathways for tumor cell survival act as a mutually interdependent network via extensive cross-talks and feedback loops. Thus, dysregulations of multiple signaling pathways usually join forces to drive oncogenesis, tumor progression, invasion, metastasis, and drug resistance, thereby providing a basis for so-called "bypass" mechanisms underlying non-genetic resistance in response to targeted agents. In this context, simultaneous interruption of two or more related targets or pathways (an approach called dual-targeted therapy, DTT), via either linear or parallel inhibition, is required to deal with such a form of drug resistance to targeted agents that specifically inhibit a single oncoprotein or oncogenic pathway. Together, while most types of tumor cells are often addicted to two or more targets or pathways or can switch their dependency between them, DTT targeting either intrinsically activated or drug-induced compensatory targets/pathways would efficiently overcome drug resistance caused by non-genetic events, with a great opportunity that those resistant cells might be particularly more vulnerable. In this review article, we discuss, with our experience, diverse mechanisms for non-genetic resistance to targeted agents and the rationales to circumvent them in the treatment of cancer, emphasizing hematologic malignancies.
Collapse
Affiliation(s)
- Wei Wang
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, China
| | - Yue Sun
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, China
| | - Xiaobo Liu
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, China
| | - Shaji K. Kumar
- Division of Hematology, Mayo Clinic College of Medicine, Rochester, MN, United States
| | - Fengyan Jin
- Department of Hematology, The First Hospital of Jilin University, Changchun, China
| | - Yun Dai
- Laboratory of Cancer Precision Medicine, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
6
|
Luo H, Song B, Xiong G, Zhang B, Zuo Z, Zhou Z, Chang X. Cadmium inhibits neural stem/progenitor cells proliferation via MitoROS-dependent AKT/GSK-3β/β-catenin signaling pathway. J Appl Toxicol 2021; 41:1998-2010. [PMID: 33977565 DOI: 10.1002/jat.4179] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2021] [Revised: 04/20/2021] [Accepted: 04/22/2021] [Indexed: 01/18/2023]
Abstract
Cadmium (Cd) is a toxic heavy metal widely found in the environment. Cd is also a potential neurotoxicant, and its exposure is associated with impairment of cognitive function. However, the underlying mechanisms by which Cd induces neurotoxicity are unclear. In this study, we investigated the in vitro effect of Cd on primary murine neural stem/progenitor cells (mNS/PCs) isolated from the subventricular zone. Our results show that Cd exposure leads to mNS/PCs G1/S arrest, promotes cell apoptosis, and inhibits cell proliferation. In addition, Cd increases intracellular and mitochondrial reactive oxygen species (ROS) that activates mitochondrial oxidative stress, decreases ATP production, and increases mitochondrial proton leak and glycolysis rate in a dose-dependent manner. Furthermore, Cd exposure decreases phosphorylation of protein kinase B (AKT) and glycogen synthase kinase-3 beta (GSK3β) in mNS/PCs. In addition, pretreatment mNS/PCs with MitoTEMPO, a mitochondrial-targeted antioxidant, improves mitochondrial morphology and functions and attenuates Cd-induced inhibition of mNS/PCs proliferation. It also effectively reverses Cd-induced changes of phosphorylation of AKT and the expression of β-catenin and its downstream genes. Taken together, our data suggested that AKT/GSK3β/β-catenin signaling pathway is involved in Cd-induced mNS/PCs proliferation inhibition via MitoROS-dependent pattern.
Collapse
Affiliation(s)
- Huan Luo
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
| | - Bo Song
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
| | - Guiya Xiong
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
| | - Bing Zhang
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
| | - Zhenzi Zuo
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
| | - Zhijun Zhou
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
| | - Xiuli Chang
- School of Public Health and Key Laboratory of Public Health Safety of the Ministry of Education, Fudan University, Shanghai, China
| |
Collapse
|
7
|
Prajumwongs P, Waenphimai O, Vaeteewoottacharn K, Wongkham S, Sawanyawisuth K. Reversine, a selective MPS1 inhibitor, induced autophagic cell death via diminished glucose uptake and ATP production in cholangiocarcinoma cells. PeerJ 2021; 9:e10637. [PMID: 33505802 PMCID: PMC7797171 DOI: 10.7717/peerj.10637] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/08/2020] [Accepted: 12/02/2020] [Indexed: 12/18/2022] Open
Abstract
Reversine is a selective inhibitor of mitotic kinase monopolar spindle 1 (MPS1) and has been reported as an anticancer agent in various cancers. The effects of reversine on bile duct cancer, cholangiocarcinoma (CCA), a lethal cancer in Northeastern Thailand, were investigated. This study reports that reversine inhibited cell proliferation of CCA cell lines in dose- and time-dependent manners but had less inhibitory effect on an immortalized cholangiocyte cell line. Reversine also triggered apoptotic cell death by decreasing anti-apoptotic proteins, Bcl-XL and Mcl-1, increasing Bax pro-apoptotic protein and activating caspase-3 activity. Moreover, reversine induced autophagic cell death by increasing LC3-II and Beclin 1 while decreasing p62. Reversine activated autophagy via the AKT signaling pathway. Additionally, this study demonstrated for the first time that reversine could diminish the expression of Hypoxia-Inducible Factor 1- alpha (HIF-1α) and glucose transporter 1 (GLUT1), resulting in a reduction of glucose uptake and energy production in CCA cell lines. These findings suggest that reversine could be a good candidate as an alternative or supplementary drug for CCA treatment.
Collapse
Affiliation(s)
- Piya Prajumwongs
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Orawan Waenphimai
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Kulthida Vaeteewoottacharn
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Sopit Wongkham
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| | - Kanlayanee Sawanyawisuth
- Department of Biochemistry, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand.,Cholangiocarcinoma Research Institute, Faculty of Medicine, Khon Kaen University, Khon Kaen, Thailand
| |
Collapse
|
8
|
Li L, Wang F, Zhang J, Wang K, De X, Li L, Zhang Y. Typical phthalic acid esters induce apoptosis by regulating the PI3K/Akt/Bcl-2 signaling pathway in rat insulinoma cells. ECOTOXICOLOGY AND ENVIRONMENTAL SAFETY 2021; 208:111461. [PMID: 33091774 DOI: 10.1016/j.ecoenv.2020.111461] [Citation(s) in RCA: 37] [Impact Index Per Article: 9.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 04/12/2020] [Revised: 07/29/2020] [Accepted: 10/01/2020] [Indexed: 06/11/2023]
Abstract
Di-(2-ethylhexyl) phthalate (DEHP) and dibutyl phthalate (DBP) are representative phthalic acid esters (PAEs), a class of environmental endocrine disruptors used as plasticizers. PAEs exposure is associated with glucose metabolism, insulin resistance, and glucose tolerance; however, the mechanism and various PAE effects on human glucose metabolism remain largely unknown. In this study, we investigated the effects of DEHP, DBP, and their mixture on rat insulinoma (INS-1) cell apoptosis and the mechanism involved in vitro. The INS-1 cells were cultured in RPMI-1640 + 10% fetal bovine serum for 24 h and pretreated with dimethyl sulfoxide (vehicle, <0.1%), DEHP (30 μM), DBP (30 μM), and their mixture (30 μM DEHP + 30 μM DBP). The methyl-thiazolyl tetrazolium bromide test was used to measure cell viability. Hoechst 33342/propidium iodide (PI) staining and Annexin V-FITC/PI staining, 2',7'-dichlorofluorescein diacetate assay, and glucose-induced insulin secretion assay were used to detect cell apoptosis rates, intracellular reactive oxygen species (ROS), and insulin secretion in INS-1, respectively. The mRNA expression levels of Bcl-2, Bax, Caspase 9, Caspase 8, Caspase 3, phosphoinositide 3-kinase (PI3K), and Akt were detected using real-time quantitative reverse transcription PCR; their protein expression levels were detected using western blotting. To the best of our knowledge, this study was the first to show that the combined effect of the two PAEs promotes a ROS-mediated PI3K/Akt/Bcl-2 pathway-induced pancreatic β cell apoptosis that is significantly higher than the effects of each PAE. Thus, safety standards and studies do not consider this effect as a significant oversight when blending PAEs. We assert that this must be addressed and corrected for establishing more impactful and safer standards.
Collapse
Affiliation(s)
- Liping Li
- School of Public Health and Management, Ningxia Medical University, Yinchuan 750004, Ningxia, China; Key Laboratory of Fertility Preservation and Maintenance of the Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia, China.
| | - Faxuan Wang
- School of Public Health and Management, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Jianjun Zhang
- Department of Epidemiology, Richard M. Fairbanks School of Public Health, Indiana University, Indianapolis, Indiana, United States.
| | - Kai Wang
- School of Public Health and Management, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Xiaoming De
- School of Public Health and Management, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Ling Li
- School of Public Health and Management, Ningxia Medical University, Yinchuan 750004, Ningxia, China.
| | - Yuhong Zhang
- School of Public Health and Management, Ningxia Medical University, Yinchuan 750004, Ningxia, China; Key Laboratory of Fertility Preservation and Maintenance of the Ministry of Education, Ningxia Medical University, Yinchuan, Ningxia, China.
| |
Collapse
|
9
|
Levobupivacaine inhibits proliferation and promotes apoptosis of breast cancer cells by suppressing the PI3K/Akt/mTOR signalling pathway. BMC Res Notes 2020; 13:386. [PMID: 32807213 PMCID: PMC7430121 DOI: 10.1186/s13104-020-05191-2] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/09/2020] [Accepted: 07/16/2020] [Indexed: 12/16/2022] Open
Abstract
Objective This study aimed to test the hypothesis that levobupivacaine has anti-tumour effects on breast cancer cells. Results Colony formation and transwell assay were used to determine breast cancer cells proliferation. Flow Cytometry (annexin V and PI staining) was used to investigate breast cancer cells apoptosis. The effects of levobupivacaine on cellular signalling and molecular response were studied with Quantitative Polymerase Chain Reaction and western blot. Induction of apoptosis was confirmed by cell viability, morphological changes showed cell shrinkage, rounding, and detachments from plates. The results of the western blot and Quantitative Polymerase Chain Reaction indicated activation of active caspase-3 and inhibition of FOXO1. The results of the flow Cytometry confirmed that levobupivacaine inhibited breast cancer cell proliferation and enhanced apoptosis of breast cancer cells. Quantitative Polymerase Chain Reaction and Western blot analysis showed increased p21 and decreased cyclin D. Quantitative Polymerase Chain Reaction and western blot analysis showed that levobupivacaine significantly increased Bax expression, accompanied by a significant decreased Bcl-2 expression and inhibition of PI3K/Akt/mTOR signalling pathway. These findings suggested that levobupivacaine inhibits proliferation and promotes breast cancer cells apoptosis in vitro.
Collapse
|
10
|
Ding YY, Wang FF, Jiang YG, Sheng YJ, Jiang MQ, Zhu X, Shi YH, Le GW. Dityrosine suppresses the cytoprotective action of thyroid hormone T3 via inhibiting thyroid hormone receptor-mediated transcriptional activation. RSC Adv 2020; 10:21057-21070. [PMID: 35518765 PMCID: PMC9054395 DOI: 10.1039/d0ra00276c] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2020] [Accepted: 05/18/2020] [Indexed: 12/25/2022] Open
Abstract
Dityrosine (Dityr) is the most common oxidized form of tyrosine. In the previous studies of mice treated with dityrosine, cell death in the pancreas, kidneys, and liver was detected in the presence of enhanced plasma triiodothyronine (T3) content. Due to its structural similarity with the thyroid hormone T3, we hypothesized that dityrosine might disrupt T3-dependent endocrine signaling. The cytotoxic effect of dityrosine was studied in C57BL/6 mice by gavage with a dityrosine dose of 320 μg per kg per day for 10 weeks. Cell death in the liver was detected in the presence of enhanced plasma thyroid hormone content in mice treated with dityrosine. The antagonistic effect of dityrosine on T3 biofunction was studied using HepG2 cells. Dityrosine incubation reduced T3 transport ability and attenuated the T3-mediated cell survival via regulation of the PI3k/Akt/MAPK pathway. Furthermore, dityrosine inhibited T3 binding to thyroid hormone receptors (TRs) and suppressed the TR-mediated transcription. Dityrosine also downregulated the expressions of T3 action-related factors. Taken together, this study demonstrates that dityrosine inhibits T3-dependent cytoprotection by competitive inhibition, resulting in downstream gene suppression. Our findings offer insights into how dityrosine acts as an antagonist of T3. These findings shed new light on cellular processes underlying the energy metabolism disorder caused by dietary oxidized protein, thus contributing to a better understanding of the diet-health axis at a cellular level.
Collapse
Affiliation(s)
- Yin-Yi Ding
- Collage of Food Science and Biotechnology, Zhejiang Gongshang University No.18, Xuezheng Street Hangzhou 310018 China +86 571-28877777 +86 571-28877777
- Food Nutrition Science Centre, Zhejiang Gongshang University Hangzhou 310018 China
| | - Fang-Fang Wang
- School of Life Science, Linyi University Linyi 276000 China
| | - Yu-Ge Jiang
- The State Key Laboratory of Food Science and Technology, Jiangnan University Wuxi 214122 China
- Center of Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University Wuxi 214122 China
| | - Yi-Jing Sheng
- Collage of Food Science and Biotechnology, Zhejiang Gongshang University No.18, Xuezheng Street Hangzhou 310018 China +86 571-28877777 +86 571-28877777
| | - Meng-Qi Jiang
- Collage of Food Science and Biotechnology, Zhejiang Gongshang University No.18, Xuezheng Street Hangzhou 310018 China +86 571-28877777 +86 571-28877777
| | - Xuan Zhu
- Collage of Food Science and Biotechnology, Zhejiang Gongshang University No.18, Xuezheng Street Hangzhou 310018 China +86 571-28877777 +86 571-28877777
| | - Yong-Hui Shi
- The State Key Laboratory of Food Science and Technology, Jiangnan University Wuxi 214122 China
- Center of Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University Wuxi 214122 China
| | - Guo-Wei Le
- The State Key Laboratory of Food Science and Technology, Jiangnan University Wuxi 214122 China
- Center of Food Nutrition and Functional Food Engineering, School of Food Science and Technology, Jiangnan University Wuxi 214122 China
| |
Collapse
|
11
|
Baruah T, Hauneihkim K, Kma L. Naringenin sensitizes lung cancer NCI-H23 cells to radiation by downregulation of akt expression and metastasis while promoting apoptosis. Pharmacogn Mag 2020. [DOI: 10.4103/pm.pm_535_19] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/04/2022] Open
|
12
|
Chowdhury HM, Sharmin N, Yuzbasioglu Baran M, Long L, Morrell NW, Trembath RC, Nasim MT. BMPRII deficiency impairs apoptosis via the BMPRII-ALK1-BclX-mediated pathway in pulmonary arterial hypertension. Hum Mol Genet 2019; 28:2161-2173. [PMID: 30809644 DOI: 10.1093/hmg/ddz047] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2019] [Revised: 02/15/2019] [Accepted: 02/18/2019] [Indexed: 02/02/2023] Open
Abstract
Pulmonary arterial hypertension (PAH) is a devastating cardiovascular disorder characterized by the remodelling of pre-capillary pulmonary arteries. The vascular remodelling observed in PAH patients results from excessive proliferation and apoptosis resistance of pulmonary arterial smooth muscle cells (PASMCs) and pulmonary arterial endothelial cells (PAECs). We have previously demonstrated that mutations in the type II receptor for bone morphogenetic protein (BMPRII) underlie the majority of the familial and inherited forms of the disease. We have further demonstrated that BMPRII deficiency promotes excessive proliferation and attenuates apoptosis in PASMCs, but the underlying mechanisms remain unclear. The major objective of this study is to investigate how BMPRII deficiency impairs apoptosis in PAH. Using multidisciplinary approaches, we demonstrate that deficiency in the expression of BMPRII impairs apoptosis by modulating the alternative splicing of the apoptotic regulator, B-cell lymphoma X (Bcl-x) transcripts: a finding observed in circulating leukocytes and lungs of PAH subjects, hypoxia-induced PAH rat lungs as well as in PASMCs and PAECs. BMPRII deficiency elicits cell specific effects: promoting the expression of Bcl-xL transcripts in PASMCs while inhibiting it in ECs, thus exerting differential apoptotic effects in these cells. The pro-survival effect of BMPRII receptor is mediated through the activin receptor-like kinase 1 (ALK1) but not the ALK3 receptor. Finally, we show that BMPRII interacts with the ALK1 receptor and pathogenic mutations in the BMPR2 gene abolish this interaction. Taken together, dysfunctional BMPRII responsiveness impairs apoptosis via the BMPRII-ALK1-Bcl-xL pathway in PAH. We suggest Bcl-xL as a potential biomarker and druggable target.
Collapse
Affiliation(s)
- H M Chowdhury
- Department of Medical and Molecular Genetics, King's College London, London, United Kingdom
| | - N Sharmin
- School of Pharmacy and Medical Sciences, University of Bradford, Bradford, United Kingdom.,Department of Pharmaceutical Technology, University of Dhaka, Dhaka, Bangladesh
| | - Merve Yuzbasioglu Baran
- School of Pharmacy and Medical Sciences, University of Bradford, Bradford, United Kingdom.,Department of Pharmacognosy, Faculty of Pharmacy, Hacettepe University, Ankara, Turkey
| | - L Long
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - N W Morrell
- Division of Respiratory Medicine, Department of Medicine, University of Cambridge, Cambridge, United Kingdom
| | - R C Trembath
- Department of Medical and Molecular Genetics, King's College London, London, United Kingdom.,National Institute for Health Research (NIHR), Biomedical Research Centre, Guy's and St. Thomas' NHS Foundation Trust and King's College London, London, United Kingdom
| | - Md Talat Nasim
- Department of Medical and Molecular Genetics, King's College London, London, United Kingdom.,School of Pharmacy and Medical Sciences, University of Bradford, Bradford, United Kingdom.,National Institute for Health Research (NIHR), Biomedical Research Centre, Guy's and St. Thomas' NHS Foundation Trust and King's College London, London, United Kingdom.,Centre for Health Agricultural and Socio-economic Advancements (CHASA), Lalmonirhat, Bangladesh
| |
Collapse
|
13
|
Anti-Remodeling Effects of Xanthohumol-Fortified Beer in Pulmonary Arterial Hypertension Mediated by ERK and AKT Inhibition. Nutrients 2019; 11:nu11030583. [PMID: 30857304 PMCID: PMC6472147 DOI: 10.3390/nu11030583] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/23/2019] [Revised: 02/27/2019] [Accepted: 02/28/2019] [Indexed: 12/17/2022] Open
Abstract
Polyphenols present in some alcoholic beverages have been linked to beneficial effects in preventing cardiovascular diseases. Polyphenols found in beer with anti-proliferative and anti-cancer properties are appealing in the context of the quasi-malignant phenotype of pulmonary arterial hypertension (PAH). Our purpose was to evaluate if the chronic ingestion of a xanthohumol-fortified beer (FB) would be able to modulate the pathophysiology of experimental PAH. Male Wistar rats with monocrotaline (MCT)-induced PAH (60 mg/kg) were allowed to drink either xanthohumol-fortified beer (MCT + FB) or 5.2% ethanol (MCT + SHAM) for a period 4 weeks. At the end of the protocol, cardiopulmonary exercise testing and hemodynamic recordings were performed, followed by sample collection for further analysis. FB intake resulted in a significant attenuation of the pulmonary vascular remodeling in MCT + FB animals. This improvement was paralleled with the downregulation in expression of proteins responsible for proliferation (ERK1/2), cell viability (AKT), and apoptosis (BCL-XL). Moreover, MCT + FB animals presented improved right ventricle (RV) function and remodeling accompanied by VEGFR-2 pathway downregulation. The present study demonstrates that a regular consumption of xanthohumol through FB modulates major remodeling pathways activated in experimental PAH.
Collapse
|
14
|
Lehman JM, Hoeksema MD, Staub J, Qian J, Harris B, Callison JC, Miao J, Shi C, Eisenberg R, Chen H, Chen SC, Massion PP. Somatostatin receptor 2 signaling promotes growth and tumor survival in small-cell lung cancer. Int J Cancer 2018; 144:1104-1114. [PMID: 30152518 DOI: 10.1002/ijc.31771] [Citation(s) in RCA: 24] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/25/2017] [Revised: 06/18/2018] [Accepted: 06/25/2018] [Indexed: 12/12/2022]
Abstract
Somatostatin receptor 2 (SSTR2) is overexpressed in a majority of neuroendocrine neoplasms, including small-cell lung carcinomas (SCLCs). SSTR2 was previously considered an inhibitory receptor on cell growth, but its agonists had poor clinical responses in multiple clinical trials. The role of this receptor as a potential therapeutic target in lung cancer merits further investigation. We evaluated the expression of SSTR2 in a cohort of 96 primary tumors from patients with SCLC and found 48% expressed SSTR2. Correlation analysis in both CCLE and an SCLC RNAseq cohort confirmed high-level expression and identified an association between NEUROD1 and SSTR2. There was a significant association with SSTR2 expression profile and poor clinical outcome. We tested whether SSTR2 expression might contribute to tumor progression through activation of downstream signaling pathways, using in vitro and in vivo systems and downregulated SSTR2 expression in lung cancer cells by shRNA. SSTR2 downregulation led to increased apoptosis and dramatically decreased tumor growth in vitro and in vivo in multiple cell lines with decreased AMPKα phosphorylation and increased oxidative metabolism. These results demonstrate a role for SSTR2 signaling in SCLC and suggest that SSTR2 is a poor prognostic biomarker in SCLC and potential future therapeutic signaling target.
Collapse
Affiliation(s)
- Jonathan M Lehman
- Division of Medical Oncology, Vanderbilt Ingram Cancer Center, Nashville, TN.,Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Nashville, TN.,Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Megan D Hoeksema
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt Ingram Cancer Center, Nashville, TN.,Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Nashville, TN
| | - Jeremy Staub
- Division of Medical Oncology, Vanderbilt Ingram Cancer Center, Nashville, TN
| | - Jun Qian
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt Ingram Cancer Center, Nashville, TN.,Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Nashville, TN
| | - Bradford Harris
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt Ingram Cancer Center, Nashville, TN
| | - J Clay Callison
- University of Tennessee Graduate School of Medicine, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Jennifer Miao
- Vanderbilt University School of Medicine, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Chanjuan Shi
- Department of Pathology, Microbiology and Immunology, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Rosana Eisenberg
- Department of Pathology, Microbiology and Immunology, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Heidi Chen
- Vanderbilt University Department of Biostatistics, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Sheau-Chiann Chen
- Vanderbilt University Department of Biostatistics, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| | - Pierre P Massion
- Division of Allergy, Pulmonary and Critical Care Medicine, Department of Medicine, Vanderbilt Ingram Cancer Center, Nashville, TN.,Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Nashville, TN.,Veterans Affairs, Tennessee Valley Healthcare System, Nashville Campus, Nashville, TN
| |
Collapse
|
15
|
Chen YC, Cheng CY, Liu CT, Sue YM, Chen TH, Hsu YH, Hwang PA, Chen CH. Alleviative effect of fucoxanthin-containing extract from brown seaweed Laminaria japonica on renal tubular cell apoptosis through upregulating Na +/H + exchanger NHE1 in chronic kidney disease mice. JOURNAL OF ETHNOPHARMACOLOGY 2018; 224:391-399. [PMID: 29920359 DOI: 10.1016/j.jep.2018.06.023] [Citation(s) in RCA: 9] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/14/2018] [Revised: 06/11/2018] [Accepted: 06/15/2018] [Indexed: 06/08/2023]
Abstract
ETHNOPHARMACOLOGICAL RELEVANCE Brown seaweed is a common food for Asians, and the bioactive ingredient fucoxanthin exerts anti-apoptotic activities in several cell types. Renal tubular cell apoptosis is one of the common cellular events leading to renal fibrosis and chronic kidney disease (CKD). However, the influence of fucoxanthin-containing brown seaweed extract on CKD is still unknown. We intended to evaluate the inhibitory effect of fucoxanthin-containing extract from brown seaweed on renal apoptosis under CKD condition and its molecular mechanism. MATERIALS AND METHODS The fucoxanthin-containing brown seaweed extract (LJE) was prepared from Laminaria japonica. We investigated how LJE influences on both doxorubicin-treated rat renal tubular cells (NRK-52E) and the renal symptoms of nephrectomy-induced CKD mice. RESULTS LJE inhibited doxorubicin-induced apoptosis and upregulated Na+/H+ exchanger isoform 1 (NHE1) expression in NRK-52E cells, which were blocked by the NHE1 inhibitor cariporide. LJE also upregulated peroxisome proliferator-activated receptor alpha (PPARα). PPARα siRNA transfection inhibited LJE-induced NHE1 expression and anti-apoptotic effect. In CKD mice, LJE increased NHE1 expression in renal tubules and reduced apoptotic renal tubular cells, but not in PPARα knockout mice. The inhibitory effect of LJE on apoptosis also reduced renal tubulointerstitial fibrosis and improved renal function in CKD mice. CONCLUSION We demonstrated that LJE inhibits renal apoptosis via NHE1 upregulation. The anti-apoptotic effect of LJE also improves renal function in CKD mice. Therefore, fucoxanthin-containing brown seaweed may have a therapeutic potential for CKD patients.
Collapse
Affiliation(s)
- Yen-Cheng Chen
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chung-Yi Cheng
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Chung-Te Liu
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yuh-Mou Sue
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Tso-Hsiao Chen
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| | - Yung-Ho Hsu
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan
| | - Pai-An Hwang
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Taiwan
| | - Cheng-Hsien Chen
- Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan; Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan; Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, New Taipei City, Taiwan.
| |
Collapse
|
16
|
Cheng X, Yu D, Cheng G, Yung BC, Liu Y, Li H, Kang C, Fang X, Tian S, Zhou X, Liu Q, Lee RJ. T7 Peptide-Conjugated Lipid Nanoparticles for Dual Modulation of Bcl-2 and Akt-1 in Lung and Cervical Carcinomas. Mol Pharm 2018; 15:4722-4732. [PMID: 30138565 DOI: 10.1021/acs.molpharmaceut.8b00696] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Expression of Bcl-2 and Akt-1 has been associated with human cancer. G3139 and RX-0201, targeting Bcl-2 and Akt-1, respectively, are antisense oligonucleotides (ASOs) that have shown limited efficacy in clinical trials. Herein, we report a combination of newly designed ASOs based on these agents and was delivered by tumor cell-targeting lipid nanoparticles (LNPs). A "Gapmer" design strategy was applied to these ASOs with the addition of 2'-O-methyl modifications on the nucleotides at 5' and 3' ends. A dual-channel syringe pump-based system was developed for the synthesis of the LNPs. ASO-LNPs composed of DODMA, egg PC, cholesterol, T7-PEG-DSPE, and PEG-DMG at a molar ratio of 35:39.5:20:0.5:5 and carrying either individual ASOs or co-loaded ASO combinations (Co-ASOs) were synthesized and evaluated in both KB and A549 cancer cells and in an A549 murine xenograft model to determine their antitumor effects and biological activities. The ASO-LNPs exhibited excellent colloidal stability and high ASO encapsulation efficiency with relatively small mean particle sizes and moderately positive zeta potentials. Transferrin receptor-targeting T7-conjugated LNPs showed enhanced cellular uptake compared to nontargeted LNPs. In addition, both T7-conjugated Co-ASOs-LNPs and non-T7-conjugated Co-ASOs-LNPs at a molar ratio of (G3139-GAP to RX-0201-GAP at 1:2) showed efficient downregulation of both Bcl-2 and Akt-1 in both A549 and KB cells. Furthermore, T7-conjugated Co-ASOs-LNPs (Co-ASOs-LNPs) produced superior antitumor activity, prolonged the overall survival time, and demonstrated tumor targeting activity in an A549 xenograft model.
Collapse
Affiliation(s)
| | - Daorui Yu
- Department of Pharmacology, School of Basic Medicine and Life Science , Hainan Medical University , Haikou , China
| | - Guang Cheng
- State Key Laboratory of Long-Acting and Targeted Drug Delivery, Nanjing , China.,Luye Sike Pharma, Nanjing Hightech Industrial Development Zone, Nanjing , China
| | - Bryant C Yung
- The WhiteOak Group, LLC., Washington , D.C. 20006 , United States
| | | | | | - Chen Kang
- Department of Internal Medicine, Division of Cardiovascular Medicine, Carver College of Medicine , University of Iowa , Iowa City , Iowa 52242 , United States
| | - Xingyue Fang
- Department of Pharmacology, School of Basic Medicine and Life Science , Hainan Medical University , Haikou , China
| | - Shuhong Tian
- Department of Pharmacology, School of Basic Medicine and Life Science , Hainan Medical University , Haikou , China
| | - Xiaoju Zhou
- School of Pharmaceutical Science , Wuhan University , Wuhan 430071 , P.R. China
| | - Qibing Liu
- Department of Pharmacology, School of Basic Medicine and Life Science , Hainan Medical University , Haikou , China
| | | |
Collapse
|
17
|
Cai Q, Lin J, Zhang L, Lin S, Peng J. Chloroform fraction of Serratulae chinensis S. Moore suppresses proliferation and induces apoptosis via the phosphatidylinositide 3-kinase/Akt pathway in human gastric cancer cells. Oncol Lett 2018; 15:8871-8877. [PMID: 29928328 DOI: 10.3892/ol.2018.8366] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2016] [Accepted: 08/03/2017] [Indexed: 11/05/2022] Open
Abstract
The chloroform fraction of the folk Chinese medicine, Serratulae chinensis S. Moore (CSC) and its anti-inflammatory activity is well recognized. However, the molecular mechanisms underlying the beneficial anticancer effects of CSC remain largely unknown. The aim of the present study was to examine the effects of CSC on the regulation of cell proliferation and apoptosis in SGC-7901 gastric cancer cells, as well as to investigate the underlying molecular mechanisms involved. The results from the present study demonstrated that CSC treatment inhibited SGC-7901 cell viability and survival in a dose- and/or time-dependent manner. CSC treatment further induced the apoptosis of SGC-7901 cells, characterized by distinct chromatin condensation and fragmented nuclear morphology. In addition, CSC treatment suppressed protein kinase-B (Akt) phosphorylation and phosphatidylinositide 3-kinase (PI3K) expression in SGC-7901 cells, which in turn promoted cancer cell apoptosis and inhibited cell proliferation. Furthermore, CSC treatment altered the expression pattern of several key target genes of the PI3K/Akt signaling pathway through the downregulation of Cyclin D1, cyclin-dependent kinase-4 and B-cell lymphoma-2 and the upregulation of Bcl-2-associated X protein. Therefore, the results from the present study demonstrated that CSC suppressed cell survival and induced apoptosis in human gastric cancer cells, via targeting the PI3K/Akt pathway.
Collapse
Affiliation(s)
- Qiaoyan Cai
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jing Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Ling Zhang
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Shan Lin
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| | - Jun Peng
- Academy of Integrative Medicine, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China.,Fujian Key Laboratory of Integrative Medicine on Geriatrics, Fujian University of Traditional Chinese Medicine, Fuzhou, Fujian 350122, P.R. China
| |
Collapse
|
18
|
Ji X, Qian J, Rahman SMJ, Siska PJ, Zou Y, Harris BK, Hoeksema MD, Trenary IA, Heidi C, Eisenberg R, Rathmell JC, Young JD, Massion PP. xCT (SLC7A11)-mediated metabolic reprogramming promotes non-small cell lung cancer progression. Oncogene 2018; 37:5007-5019. [PMID: 29789716 PMCID: PMC6127081 DOI: 10.1038/s41388-018-0307-z] [Citation(s) in RCA: 222] [Impact Index Per Article: 31.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 03/23/2018] [Accepted: 04/13/2018] [Indexed: 01/10/2023]
Abstract
Many tumors increase uptake and dependence on glucose, cystine or glutamine. These basic observations on cancer cell metabolism have opened multiple new diagnostic and therapeutic avenues in cancer research. Recent studies demonstrated that smoking could induce the expression of xCT (SLC7A11) in oral cancer cells, suggesting that overexpression of xCT may support lung tumor progression. We hypothesized that overexpression of xCT occurs in lung cancer cells to satisfy the metabolic requirements for growth and survival. Our results demonstrated that 1) xCT was highly expressed at the cytoplasmic membrane in non-small cell lung cancer (NSCLC), 2) the expression of xCT was correlated with advanced stage and predicted a worse 5-year survival, 3) targeting xCT transport activity in xCT overexpressing NSCLC cells with sulfasalazine decreased cell proliferation and invasion in vitro and in vivo and 4) increased dependence on glutamine was observed in xCT overexpressed normal airway epithelial cells. These results suggested that xCT regulate metabolic requirements during lung cancer progression and be a potential therapeutic target in NSCLC.
Collapse
Affiliation(s)
- Xiangming Ji
- Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA.,Department of Nutrition, Byrdine F. Lewis School of Nursing and Health Professions, Georgia State University, Atlanta, 30302, USA
| | - Jun Qian
- Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - S M Jamshedur Rahman
- Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Peter J Siska
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Yong Zou
- Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Bradford K Harris
- Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Megan D Hoeksema
- Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Irina A Trenary
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, USA
| | - Chen Heidi
- Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA
| | - Rosana Eisenberg
- Department of Pathology, Microbiology, and Immunology, Vanderbilt University School of Medicine, Nashville, USA
| | - Jeffrey C Rathmell
- Department of Internal Medicine III, University Hospital Regensburg, 93053, Regensburg, Germany
| | - Jamey D Young
- Department of Chemical and Biomolecular Engineering, Vanderbilt University, Nashville, USA
| | - Pierre P Massion
- Cancer Early Detection and Prevention Initiative, Vanderbilt Ingram Cancer Center, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine, Nashville, TN, 37232, USA. .,Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN, 37212, USA.
| |
Collapse
|
19
|
Davis JB, Calvert V, Roberts S, Bracero S, Petricoin E, Couch R. Induction of nerve growth factor by phorbol 12-myristate 13-acetate is dependent upon the mitogen activated protein kinase pathway. Heliyon 2018; 4:e00617. [PMID: 29872754 PMCID: PMC5986306 DOI: 10.1016/j.heliyon.2018.e00617] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/09/2017] [Revised: 01/23/2018] [Accepted: 04/27/2018] [Indexed: 01/20/2023] Open
Abstract
Several small molecules have been identified that induce glial cells to synthesize and secrete nerve growth factor (NGF), a critical neurotrophin that supports neuronal growth and survival, and as such show promise in the development of drugs for the chemoprevention of Alzheimer's disease. To map the signal transduction cascade leading to NGF synthesis and secretion, cultured human glial cells were stimulated by phorbol 12-myristate 13-acetate (PMA), an agonist of Protein Kinase C. Changes in intracellular protein phosphorylation states were evaluated by reverse phase protein microarrays (RPPA), selectively screening over 130 protein endpoints. Of these, 55 proteins showed statistically significant changes in phosphorylation state due to cellular exposure to PMA. A critical signal transduction pathway was identified, and subsequent validation by ELISA and qPCR revealed that the signaling proteins Raf, MEK, ERK, and the signal transduction factor CREB are all essential to the upregulation of NGF gene expression by PMA. Additionally, members of the RSK family of kinases appear to be involved in glial secretion (exocytosis) of the NGF protein. Furthermore, through RPPA, the effects of PMA on apoptosis signaling events and cell proliferation were differentiated from the pathway to NGF upregulation. Overall, this study reveals potential protein targets for the rational design of Alzheimer's therapeutics.
Collapse
|
20
|
Alotaibi MR, Hassan ZK, Al-Rejaie SS, Alshammari MA, Almutairi MM, Alhoshani AR, Alanazi WA, Hafez MM, Al-Shabanah OA. Characterization of Apoptosis in a Breast Cancer Cell Line after IL-10 Silencing. Asian Pac J Cancer Prev 2018; 19:777-783. [PMID: 29582634 PMCID: PMC5980855 DOI: 10.22034/apjcp.2018.19.3.777] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Background: Breast cancer is affected by the immune system in that different cytokines play roles in its initiation
and progression. Interleukin-10 (IL-10), an anti-inflammatory cytokine, is an immunosuppressive factor involved in
tumorigenesis. The present study was conducted to investigate the gene silencing effect of a small interference RNA
(siRNA) targeting IL-10 on the apoptotic pathway in breast cancer cell line. Methods: The siRNA targeting IL-10 and
a glyceraldehyde 3-phosphate dehydrogenase (GAPDH) clone were introduced into MDA-MB-231 cells. Real-time
PCR assays were used to determine IL-10 and GAPDH gene expression levels, in addition to those for protein kinase
B (AKT), phosphoinositide 3-kinase (PI3K), B-cell lymphoma 2 (Bcl2), caspase-3 and caspase-9 genes related to
apoptosis. Results: Inhibition of IL-10 by the siRNA accelerated apoptosis and was accompanied by significant
increase in caspase-3 and caspase-9 and a significant decrease in PI3K, AKT and Bcl2 expression levels compared to
the non-transfected case. Conclusions: In conclusion, the production of IL-10 may represent a new escape mechanism
by breast cancer cells to evade destruction by the immune system. IL-10 gene silencing causes down regulation of both
PI3K/AKT and Bcl2 gene expression and also increases the Bbc3, BAX caspase3, and caspase 3 cleavage expression
levels. IL–10 might represent a promising new target for therapeutic strategies.
Collapse
Affiliation(s)
- Moureq R Alotaibi
- College of Pharmacy, Pharmacology and Toxicology Department, Kind Saud University, Riyadh, kingdom of Saudi Arabia
| | | | | | | | | | | | | | | | | |
Collapse
|
21
|
Deshpande RP, Babu PP. pDok2, caspase 3 dependent glioma cell growth arrest by nitidine chloride. Pharmacol Rep 2017; 70:48-54. [PMID: 29329030 DOI: 10.1016/j.pharep.2017.07.013] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Revised: 05/22/2017] [Accepted: 07/12/2017] [Indexed: 12/28/2022]
Abstract
BACKGROUND Nitidine chloride (NC) is known to exert anticancer and anti-metastatic effects on a variety of tumors. Recently, NC has also been shown to inhibit PIK3/AKT/mTOR axis in U87 human glioma cells. METHODS The study shows NC employing pDok2, caspase 3 dependent cell death in C6 rat glioma and U87 human malignant glioblastoma cells. The effect of NC on glioblastoma cell lines was accessed by MTT, clonogenic and wound healing assays. Cell cycle analysis was performed by FACS. Moreover, the effect of NC on downstream target proteins, such as caspase3, pDok2, PARP, and Gsk3 beta, were measured by western blotting. RESULTS Overexpressed pDok2 protein has recently been reported as a prognostic marker with poor outcomes for human glioblastoma multiformae. We found that NC inhibits pDok2 in U87 cells in a concentration-dependent way. We further showed that cleaved PARP and cleaved caspase 3 protein expressions were increased in C6 cells treated with NC in a dose-dependent way. NC effectively attenuated C6 cells growth and colony formation at 8μM (micromoles) concentration. Cell cycle arrest in G2/M phase was further confirmed by flow cytometry. NC also exhibited its inhibitory effect on Gsk3 beta, which has been proven to be altered in glioma biology. CONCLUSIONS Collectively, we predicted that NC could be employed as a potential anti-glioma mediator that needs attention to explore the mechanisms of its activity.
Collapse
Affiliation(s)
- Ravindra Pramod Deshpande
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana State, India.
| | - Phanithi Prakash Babu
- Department of Biotechnology and Bioinformatics, School of Life Sciences, University of Hyderabad, Hyderabad, Telangana State, India.
| |
Collapse
|
22
|
Liu CY, Hsieh CH, Kim SH, Wang JP, Ni YL, Su CL, Yao CF, Fang K. An indolylquinoline derivative activates DNA damage response and apoptosis in human hepatocellular carcinoma cells. Int J Oncol 2016; 49:2431-2441. [PMID: 27748837 DOI: 10.3892/ijo.2016.3717] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Accepted: 09/14/2016] [Indexed: 12/17/2022] Open
Abstract
Human liver cancer is one of the most frequently diagnosed cancers worldwide. The development of resistance to therapy limits the application against the disease. To improve treatment, new effective anticancer agents are constantly pursued. Previously, we reported that an indolylquinoline, 3-((7-ethyl-1H-indol-3-yl)-methyl)-2-methylquinoline (EMMQ), is effective in suppressing the growth of human lung cancer by impairing mitochondria functions. The present study revealed that EMMQ inhibited cell growth and induced apoptosis in liver cancer cells, but not in normal cells. This study demonstrated that EMMQ induced DNA damage by activating p53 and γ-H2AX and cell arrest by suppressing cyclin D1 and CDK2. Damaged DNA injured mitochondrial functions by lowering the membrane potential and producing reactive oxygen species. The subsequent mitochondrial cytochrome c release attenuated pro-survival signals and increased apoptotic characteristics. Introduction of p53 shRNA abrogated drug effects by reducing DNA damage while maintaining mitochondria integrity. In brief, the study demonstrates that the effectiveness of EMMQ accentuated apoptosis of hepatocarcinoma cells by activating p53. Based on these collective findings, the study offered a new perspective of EMMQ that was shown to be a promising candidate to treat liver cancer.
Collapse
Affiliation(s)
- Chun-Yen Liu
- Department of Life Science, National Taiwan Normal University, Taipei 116, Taiwan, R.O.C
| | - Chang-Hung Hsieh
- Department of Life Science, National Taiwan Normal University, Taipei 116, Taiwan, R.O.C
| | - Seung-Hun Kim
- Department of Life Science, National Taiwan Normal University, Taipei 116, Taiwan, R.O.C
| | - Jing-Ping Wang
- Department of Life Science, National Taiwan Normal University, Taipei 116, Taiwan, R.O.C
| | - Yu-Lin Ni
- Department of Life Science, National Taiwan Normal University, Taipei 116, Taiwan, R.O.C
| | - Chun-Li Su
- Department of Human Development and Family Studies, National Taiwan Normal University, Taipei 116, Taiwan, R.O.C
| | - Ching-Fa Yao
- Department of Chemistry, National Taiwan Normal University, Taipei 116, Taiwan, R.O.C
| | - Kang Fang
- Department of Life Science, National Taiwan Normal University, Taipei 116, Taiwan, R.O.C
| |
Collapse
|
23
|
Potter DS, Galvin M, Brown S, Lallo A, Hodgkinson CL, Blackhall F, Morrow CJ, Dive C. Inhibition of PI3K/BMX Cell Survival Pathway Sensitizes to BH3 Mimetics in SCLC. Mol Cancer Ther 2016; 15:1248-60. [PMID: 27197306 DOI: 10.1158/1535-7163.mct-15-0885] [Citation(s) in RCA: 28] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2015] [Accepted: 03/10/2016] [Indexed: 11/16/2022]
Abstract
Most small cell lung cancer (SCLC) patients are initially responsive to cytotoxic chemotherapy, but almost all undergo fatal relapse with progressive disease, highlighting an urgent need for improved therapies and better patient outcomes in this disease. The proapoptotic BH3 mimetic ABT-737 that targets BCL-2 family proteins demonstrated good single-agent efficacy in preclinical SCLC models. However, so far clinical trials of the BH3 mimetic Navitoclax have been disappointing. We previously demonstrated that inhibition of a PI3K/BMX cell survival signaling pathway sensitized colorectal cancer cells to ABT-737. Here, we show that SCLC cell lines, which express high levels of BMX, become sensitized to ABT-737 upon inhibition of PI3K in vitro, and this is dependent on inhibition of the PI3K-BMX-AKT/mTOR signaling pathway. Consistent with these cell line data, when combined with Navitoclax, PI3K inhibition suppressed tumor growth in both an established SCLC xenograft model and in a newly established circulating tumor cell-derived explant (CDX) model generated from a blood sample obtained at presentation from a chemorefractory SCLC patient. These data show for the first time that a PI3K/BMX signaling pathway plays a role in SCLC cell survival and that a BH3 mimetic plus PI3K inhibition causes prolonged tumor regression in a chemorefractory SCLC patient-derived model in vivo These data add to a body of evidence that this combination should move toward the clinic. Mol Cancer Ther; 15(6); 1248-60. ©2016 AACR.
Collapse
Affiliation(s)
- Danielle S Potter
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Melanie Galvin
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Stewart Brown
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Alice Lallo
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Cassandra L Hodgkinson
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Fiona Blackhall
- Institute of Cancer Sciences, University of Manchester, Manchester, United Kingdom. Christie NHS Foundation Trust, Manchester, United Kingdom
| | - Christopher J Morrow
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Caroline Dive
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom. CRUK Lung Cancer Centre of Excellence, Manchester, United Kingdom.
| |
Collapse
|
24
|
Liu CY, Wu PT, Wang JP, Fan PW, Hsieh CH, Su CL, Chiu CC, Yao CF, Fang K. An indolylquinoline derivative promotes apoptosis in human lung cancer cells by impairing mitochondrial functions. Apoptosis 2016; 20:1471-82. [PMID: 26349782 DOI: 10.1007/s10495-015-1165-6] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
A number of effective anti-cancer drugs contain either indole or quinoline group. Compounds fused indole and quinoline moieties altogether as indolylquinoline were rarely reported as anti-cancer agents. We reported here that a synthetic indolylquinoline derivative, 3-((7-ethyl-1H-indol-3-yl)-methyl)-2-methylquinoline (EMMQ), inhibited the growth of human non-small cell lung cancer (NSCLC) cells in dose- and time-dependent manners. The cytotoxicity was mediated through apoptotic cell death that began with mitochondrial membrane potential interruption and DNA damage. EMMQ caused transient elevation of p53 that assists in cytochrome c release, cleavage of downstream PARP and procaspase-3 and mitochondria-related apoptosis. The degree of apoptotic cell death depends on the status of tumor suppressor p53 of the target cells. H1299 cells with stable ectopic expression of p53 induced cytotoxicity by disrupting mitochondria functions that differed with those transfected with mutant p53. Knocking-down of p53 attenuated drug effects. EMMQ suppressed the growth of A549 tumor cells in xenograft tumors by exhibiting apoptosis characteristics. Given its small molecular weight acting as an effective p53 regulator in NSCLC cells, EMMQ could be an addition to the current list of lung cancer treatment.
Collapse
Affiliation(s)
- Chun-Yen Liu
- Department of Life Science, National Taiwan Normal University, 88 Ting-Chow Rd, Sec 4, Taipei, 116, Taiwan
| | - Pei-Tsen Wu
- Department of Life Science, National Taiwan Normal University, 88 Ting-Chow Rd, Sec 4, Taipei, 116, Taiwan
| | - Jing-Ping Wang
- Department of Life Science, National Taiwan Normal University, 88 Ting-Chow Rd, Sec 4, Taipei, 116, Taiwan
| | - Po-Wei Fan
- Department of Life Science, National Taiwan Normal University, 88 Ting-Chow Rd, Sec 4, Taipei, 116, Taiwan
| | - Chang-Hung Hsieh
- Department of Life Science, National Taiwan Normal University, 88 Ting-Chow Rd, Sec 4, Taipei, 116, Taiwan
| | - Chun-Li Su
- Department of Human Development and Family Studies, National Taiwan Normal University, Taipei, Taiwan
| | - Chien-Chih Chiu
- Department of Biotechnology, Kaohsiung Medical University, Kaohsiung, Taiwan
| | - Ching-Fa Yao
- Department of Chemistry, National Taiwan Normal University, Taipei, Taiwan
| | - Kang Fang
- Department of Life Science, National Taiwan Normal University, 88 Ting-Chow Rd, Sec 4, Taipei, 116, Taiwan.
| |
Collapse
|
25
|
Chen CH, Chen TH, Wu MY, Chen JR, Hong LY, Zheng CM, Chiu IJ, Lin YF, Hsu YH. Peroxisome Proliferator-Activated Receptor α Protects Renal Tubular Cells from Gentamicin-Induced Apoptosis via Upregulating Na +/H + Exchanger NHE1. Mol Med 2015; 21:886-889. [PMID: 26623927 DOI: 10.2119/molmed.2015.00196] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2015] [Accepted: 11/23/2015] [Indexed: 12/18/2022] Open
Abstract
Peroxisome proliferator-activated receptor (PPAR)-α is a transcription factor that has been reported to inhibit gentamicin-induced apoptosis in renal tubular cells. However, the antiapoptotic mechanism of PPARα is still unknown. In this study, we found that PPARα overexpression induced Na+/H+ exchanger-1 (NHE1) expression in the rat renal tubular cells NRK-52E. Beraprost, a PPARα ligand, also increased NHE1 expression in the renal tubules in normal mice, but not in PPARα knockout mice. Chromatin immunoprecipitation assays revealed that two PPARα binding elements were located in the rat NHE1 promoter region. Na+/H+ exchanger activity also increased in the PPARα-overexpressed cells. Flow cytometry showed that the PPARα-overexpressed cells were resistant to apoptosis-induced shrinkage. Cariporide, a selective NHE1 inhibitor, inhibited the antiapoptotic effect of PPARα in the gentamicin-treated cells. The interaction between NHE1 and ezrin/radixin/moesin (ERM) and between ERM and phosphatidylinositol 4,5-bisphosphate in the PPARα-overexpressed cells was more than in the control cells. ERM short interfering RNA (siRNA) transfection inhibited the PPARα-induced antiapoptotic effect. PPARα overexpression also increased the phosphoinositide 3-kinase (PI3K) expression, which is dependent on NHE1 activity. Increased PI3K further increased the phosphorylation of the prosurvival kinase Akt in the PPARα-overexpressed cells. Wortmannin, a PI3K inhibitor, inhibited PPARα-induced Akt activity and the antiapoptotic effect. We conclude that PPARα induces NHE1 expression and then recruits ERM to promote PI3K/Akt-mediated cell survival in renal tubular cells. The application of PPARα activation reduces the nephrotoxicity of gentamicin and may expand the clinical use of gentamicin.
Collapse
Affiliation(s)
- Cheng-Hsien Chen
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Tso-Hsiao Chen
- Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan.,Division of Nephrology, Department of Internal Medicine, Wan Fang Hospital, Taipei Medical University, Taipei, Taiwan
| | - Mei-Yi Wu
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Jia-Rung Chen
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Li-Yu Hong
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Cai-Mei Zheng
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - I-Jen Chiu
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yuh-Feng Lin
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan
| | - Yung-Ho Hsu
- Division of Nephrology, Department of Internal Medicine, Shuang Ho Hospital, Taipei Medical University, Taipei, Taiwan.,Department of Internal Medicine, School of Medicine, College of Medicine, Taipei Medical University, Taipei, Taiwan
| |
Collapse
|
26
|
Marine lipopeptide Iturin A inhibits Akt mediated GSK3β and FoxO3a signaling and triggers apoptosis in breast cancer. Sci Rep 2015; 5:10316. [PMID: 25974307 PMCID: PMC4431395 DOI: 10.1038/srep10316] [Citation(s) in RCA: 85] [Impact Index Per Article: 8.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2015] [Accepted: 03/26/2015] [Indexed: 01/18/2023] Open
Abstract
Akt kinase is a critical component of the PI3K/Akt signaling pathway, which is frequently over expressed in human cancers including breast. Therapeutic regimens for inhibiting breast cancer with aberrant Akt activity are essential. Here, we evaluated antitumor effect of a marine bacteria derived lipopeptide ‘Iturin A’ on human breast cancer in vitro and in vivo through disrupting Akt pathway. Proliferation of MDA-MB-231 and MCF-7 breast cancer cells were significantly inhibited by Iturin A and it induced apoptosis as confirmed by increased Sub G1 populations, DNA fragmentation, morphological changes and western blot analysis. Furthermore, Iturin A inhibited EGF induced Akt phosphorylation (Ser473 and Thr308) and its downstream targets GSK3β and FoxO3a. Iturin A inactivated MAPK as well as Akt kinase leading to the translocation of FoxO3a to the nucleus. Gene silencing of Akt in MDA-MB-231 and MCF-7 cells reduced the sensitivity of cancer cells to Iturin A. Interestingly, overexpression of Akt with Akt plasmid in cancer cells caused highly susceptible to induce apoptosis by Iturin A treatment. In a xenograft model, Iturin A inhibited tumor growth with reduced expressions of Ki-67, CD-31, P-Akt, P-GSK3β, P-FoxO3a and P-MAPK. Collectively, these findings imply that Iturin A has potential anticancer effect on breast cancer.
Collapse
|
27
|
Hassanein M, Qian J, Hoeksema MD, Wang J, Jacobovitz M, Ji X, Harris FT, Harris BK, Boyd KL, Chen H, Eisenberg R, Massion PP. Targeting SLC1a5-mediated glutamine dependence in non-small cell lung cancer. Int J Cancer 2015; 137:1587-97. [PMID: 25821004 DOI: 10.1002/ijc.29535] [Citation(s) in RCA: 126] [Impact Index Per Article: 12.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/17/2015] [Accepted: 03/17/2015] [Indexed: 01/22/2023]
Abstract
We previously elucidated the pleotropic role of solute carrier family A1 member 5 (SLC1A5) as the primary transporter of glutamine (Gln), a modulator of cell growth and oxidative stress in non-small cell lung cancer (NSCLC). The aim of our study was to evaluate SLC1A5 as a potential new therapeutic target and candidate biomarker predictive of survival and response to therapy. SLC1A5 targeting was examined in a panel of NSCLC and human bronchial cell lines by RNA interference and by a small molecular inhibitor, gamma-l-glutamyl-p-nitroanilide (GPNA). The effects of targeting SLC1A5 on cell growth, Gln uptake, ATP level, autophagy and cell death were examined. Inactivation of SLC1A5 genetically or pharmacologically decreased Gln consumption, inhibited cell growth, induced autophagy and apoptosis in a subgroup of NSCLC cell lines that overexpress SLC1A5. Targeting SLC1A5 function decreased tumor growth in NSCLC xenografts. A multivariate Cox proportional hazards analysis indicates that patients with increased SLC1A5 mRNA expression have significantly shorter overall survival (p = 0.01, HR = 1.24, 95% CI: 1.05-1.46), adjusted for age, gender, smoking history and disease stage. In an immunohistochemistry study on 207 NSCLC patients, SLC1A5 protein expression remained highly significant prognostic value in both univariate (p < 0.0001, HR = 1.45, 95% CI: 1.15-1.50) and multivariate analyses (p = 0.04, HR = 1.22, 95% CI: 1.01-1.31). These results position SLC1A5 as a new candidate prognostic biomarker for selective targeting of Gln-dependent NSCLC.
Collapse
Affiliation(s)
- Mohamed Hassanein
- Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center and Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, TN
| | - Jun Qian
- Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center and Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, TN
| | - Megan D Hoeksema
- Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center and Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, TN
| | - Jing Wang
- Department of Biomedical Informatics, Vanderbilt University, Nashville, TN
| | - Marie Jacobovitz
- Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center and Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, TN
| | - Xiangming Ji
- Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center and Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, TN
| | - Fredrick T Harris
- Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center and Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, TN.,Department of Biochemistry and Cancer Biology, Meharry Medical College, Nashville, TN
| | - Bradford K Harris
- Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center and Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, TN
| | - Kelli L Boyd
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN
| | - Heidi Chen
- Department of Biostatistics, Vanderbilt University, Nashville, TN
| | - Rosana Eisenberg
- Department of Pathology, Microbiology and Immunology, Vanderbilt University, Nashville, TN
| | - Pierre P Massion
- Thoracic Program, Vanderbilt-Ingram Comprehensive Cancer Center and Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University, Nashville, TN.,Veterans Affairs, Tennessee Valley Healthcare System, Nashville, TN
| |
Collapse
|
28
|
Hyun HB, Lee WS, Go SI, Nagappan A, Park C, Han MH, Hong SH, Kim G, Kim GY, Cheong J, Ryu CH, Shin SC, Choi YH. The flavonoid morin from Moraceae induces apoptosis by modulation of Bcl-2 family members and Fas receptor in HCT 116 cells. Int J Oncol 2015; 46:2670-8. [PMID: 25892545 DOI: 10.3892/ijo.2015.2967] [Citation(s) in RCA: 54] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 03/27/2015] [Indexed: 11/06/2022] Open
Abstract
It is evident based on literature that flavonoids from fruit can safely modulate cancer cell biology and induce apoptosis. Therefore, we investigated the anticancer activity of morin, a flavonoid which is plentiful in twigs of mulberry focusing on apoptosis, and its mechanisms. Morin upregulated the Fas receptor, and activates caspase-8, -9 and -3 in HCT-116 cells. Morin also activates Bid, and induced the loss of mitochondrial membrane potential (MMP, ∆Ψm) with Bax protein activation and cytochrome c release. In addition, morin induced ROS generation which was not blocked by N-acetylcysteine. Morin also suppressed Bcl-2 and cIAP-1, anti-apoptotic proteins, which may contribute to augmentation of morin-triggered apoptosis. As an upstream signaling pathway, suppressed Akt activity by morin was associated to apoptosis. This study suggests that morin induces caspase-dependent apoptosis through extrinsic pathway by upregulating Fas receptor as well as through the intrinsic pathway by modulating Bcl-2 and IAP family members, and ROS generation, and that Akt is the critical upstream signaling that regulates the apoptotic effect of morin in human colon cancer HCT-116 cells.
Collapse
Affiliation(s)
- Hwang-Bo Hyun
- Department of Biochemistry, Dongeui University College of Korean Medicine, Busan 614-052, Republic of Korea
| | - Won Sup Lee
- Department of Internal Medicine, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-702, Republic of Korea
| | - Se-Il Go
- Department of Internal Medicine, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-702, Republic of Korea
| | - Arulkumar Nagappan
- Department of Internal Medicine, Institute of Health Sciences, Gyeongsang National University School of Medicine, Jinju 660-702, Republic of Korea
| | - Cheol Park
- Department of Molecular Biology, College of Natural Sciences, Dongeui University, Busan 614-714, Republic of Korea
| | - Min Ho Han
- Department of Biochemistry, Dongeui University College of Oriental Medicine, Busan 614-052, Republic of Korea
| | - Su Hyun Hong
- Department of Biochemistry, Dongeui University College of Oriental Medicine, Busan 614-052, Republic of Korea
| | - Gonsup Kim
- School of Veterinary Medicine, Research Institute of Life Science, Gyeongsang National University, Jinju 660-701, Republic of Korea
| | - Gi Young Kim
- Laboratory of Immunobiology, Department of Marine Life Sciences, Jeju National University, Jeju 690-756, Republic of Korea
| | - Jaehun Cheong
- Department of Molecular Biology, College of Natural Sciences, Pusan National University, Busan 609-735, Republic of Korea
| | - Chung Ho Ryu
- Division of Applied Life Science (BK 21 Program), Research Institute of Life Science, Gyeongsang National University, Jinju 660-701, Republic of Korea
| | - Sung Chul Shin
- Department of Chemistry, Research Institute of Life Science, Gyeongsang National University, Jinju 660-701, Republic of Korea
| | - Yung Hyun Choi
- Department of Biochemistry, Dongeui University College of Korean Medicine, Busan 614-052, Republic of Korea
| |
Collapse
|
29
|
Kaur E, Rajendra J, Jadhav S, Shridhar E, Goda JS, Moiyadi A, Dutt S. Radiation-induced homotypic cell fusions of innately resistant glioblastoma cells mediate their sustained survival and recurrence. Carcinogenesis 2015; 36:685-95. [PMID: 25863126 DOI: 10.1093/carcin/bgv050] [Citation(s) in RCA: 60] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2014] [Accepted: 03/23/2015] [Indexed: 12/13/2022] Open
Abstract
Understanding of molecular events underlying resistance and relapse in glioblastoma (GBM) is hampered due to lack of accessibility to resistant cells from patients undergone therapy. Therefore, we mimicked clinical scenario in an in vitro cellular model developed from five GBM grade IV primary patient samples and two cell lines. We show that upon exposure to lethal dose of radiation, a subpopulation of GBM cells, innately resistant to radiation, survive and transiently arrest in G2/M phase via inhibitory pCdk1(Y15). Although arrested, these cells show multinucleated and giant cell phenotype (MNGC). Significantly, we demonstrate that these MNGCs are not pre-existing giant cells from parent population but formed via radiation-induced homotypic cell fusions among resistant cells. Furthermore, cell fusions induce senescence, high expression of senescence-associated secretory proteins (SASPs) and activation of pro-survival signals (pAKT, BIRC3 and Bcl-xL) in MNGCs. Importantly, following transient non-proliferation, MNGCs escape senescence and despite having multiple spindle poles during mitosis, they overcome mitotic catastrophe to undergo normal cytokinesis forming mononucleated relapse population. This is the first report showing radiation-induced homotypic cell fusions as novel non-genetic mechanism in radiation-resistant cells to sustain survival. These data also underscore the importance of non-proliferative phase in resistant glioma cells. Accordingly, we show that pushing resistant cells into premature mitosis by Wee1 kinase inhibitor prevents pCdk1(Y15)-mediated cell cycle arrest and relapse. Taken together, our data provide novel molecular insights into a multistep process of radiation survival and relapse in GBM that can be exploited for therapeutic interventions.
Collapse
Affiliation(s)
- Ekjot Kaur
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Jacinth Rajendra
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Shailesh Jadhav
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Epari Shridhar
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Jayant Sastri Goda
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Aliasgar Moiyadi
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| | - Shilpee Dutt
- Tata Memorial Centre, Advanced Centre for Treatment, Research and Education in Cancer, Navi Mumbai 410210, India
| |
Collapse
|
30
|
Sousa V, Bastos B, Silva M, Alarcão AM, Carvalho L. Bronchial-pulmonary adenocarcinoma subtyping relates with different molecular pathways. REVISTA PORTUGUESA DE PNEUMOLOGIA 2015; 21:259-70. [PMID: 25926247 DOI: 10.1016/j.rppnen.2014.05.006] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Accepted: 05/30/2014] [Indexed: 01/30/2023] Open
Abstract
Lung cancer is one of the most common cancers in the world with a high mortality rate. We analyzed 45 surgical samples of the adenocarcinoma, 13 with lymph node metastasis. APC, BCL2, chromogranin A, CK 5/6/18 (LP34), CK20, CK7, cyclin D1, EGFR, ERCC1, HER2, Ki67, LRP, MRP, P53, RB and TTF1 expressions were evaluated by immunohistochemistry (IHC). Higher Ki67, APC, ERCC1 expressions and lower TTF1 expression were identified in advanced stages (IIA and IIIA) of adenocarcinomas, which reflect a more aggressive, less differentiated, possibly a non-TRU adenocarcinoma. Acinar, micropapillary and BA/lepidic adenocarcinoma patterns were the most similar patterns and papillary was the most different pattern followed by solid pattern, according to expression of these markers. Different adenocarcinoma patterns are engaged with different molecular pathways for carcinogenesis, based on the differences of expression. Acinar, BA/lepidic and micropapillary showed higher TTF1 expression (type TRU), and papillary and solid patterns revealed less TTF1 expression, exhibiting a non-TRU/bronchial phenotype. Solid pattern revealed lower HER2 and higher EGFR and ERCC1 (this compared to papillary) expression; papillary higher HER2 and lower ERCC1 expressions; micropapillary higher RB expression; and acinar lower ERCC1 and higher EGFR expressions. Ciclin D1 seems to have more importance in acinar and BA/lepidic patterns than in micropapillary. ERCC1 protein expression in micropapillary, solid and BA/lepidic patterns may indicate DNA repair activation. Inhibition of apoptosis could be explained by BCL2 overexpression, present in all adenocarcinoma patterns. MRP-1 and LRP were overexpressed in all patterns, which may have implications for drug resistance. Further studies are needed to interpret these data regarding to therapy response in advanced staged bronchial-pulmonary carcinomas.
Collapse
Affiliation(s)
- Vítor Sousa
- IAP-FMUC-Institute of Pathology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Pathology Institute, Coimbra University Hospital (HUC-CHUC), Coimbra, Portugal.
| | - Bruno Bastos
- IAP-FMUC-Institute of Pathology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Maria Silva
- IAP-FMUC-Institute of Pathology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Maria Alarcão
- IAP-FMUC-Institute of Pathology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Lina Carvalho
- IAP-FMUC-Institute of Pathology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Pathology Institute, Coimbra University Hospital (HUC-CHUC), Coimbra, Portugal
| |
Collapse
|
31
|
Sun Q, Yogosawa S, Iizumi Y, Sakai T, Sowa Y. The alkaloid emetine sensitizes ovarian carcinoma cells to cisplatin through downregulation of bcl-xL. Int J Oncol 2014; 46:389-94. [PMID: 25310746 DOI: 10.3892/ijo.2014.2703] [Citation(s) in RCA: 22] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2014] [Accepted: 09/24/2014] [Indexed: 11/06/2022] Open
Abstract
Cisplatin and its platinum derivatives are first-line chemotherapeutic agents in the treatment of ovarian cancer. However, chemoresistance is the leading cause of therapeutic failure and is responsible for the poor overall survival rate. Here, we describe that emetine, a natural alkaloid used as an anti-amoebiasis drug, sensitized ovarian carcinoma cells to apoptosis induced by cisplatin. The single administration of cisplatin or emetine had a weak effect on cell death. However, co-treatment of cisplatin and emetine remarkably induced apoptosis and reduced the colony formation of ovarian carcinoma cells. Moreover, we showed that apoptosis induced by the combination of cisplatin and emetine was dependent on the activation of caspases -3, -7 and -8. As to the mechanism, downregulation of bcl-xL by emetine was shown to be responsible for enhancing the sensitivity of ovarian cancer cells to cisplatin. These findings suggest that the combination of cisplatin and emetine might be a promising treatment for ovarian cancer.
Collapse
Affiliation(s)
- Qi Sun
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Shingo Yogosawa
- Department of Public Health and Environmental Medicine, The Jikei University School of Medicine, Tokyo, Japan
| | - Yosuke Iizumi
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Toshiyuki Sakai
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| | - Yoshihiro Sowa
- Department of Molecular-Targeting Cancer Prevention, Graduate School of Medical Science, Kyoto Prefectural University of Medicine, Kyoto, Japan
| |
Collapse
|
32
|
Prasad SB, Yadav SS, Das M, Govardhan HB, Pandey LK, Singh S, Pradhan S, Narayan G. Down Regulation of FOXO1 Promotes Cell Proliferation in Cervical Cancer. J Cancer 2014; 5:655-62. [PMID: 25157276 PMCID: PMC4142327 DOI: 10.7150/jca.6554] [Citation(s) in RCA: 42] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2013] [Accepted: 07/09/2013] [Indexed: 01/17/2023] Open
Abstract
The Forkhead transcription factor FOXO1, an important downstream target of phosphatidylinositol-3-kinase (PI3K)/AKT signaling pathway, regulates cellular homeostasis by maintaining cell proliferation, apoptosis and viability in normal cells. Though, the function and regulation of FOXO1 is well documented in many cancers, the molecular mechanism of its regulation in cervical cancer is largely unknown. In the present study we have investigated the role of PI3K inhibition on FOXO1 regulation. Expression profiling of primary tumors and cell lines show over expression of PIK3CA and AKT1; and down regulation of FOXO1. Lack of FOXO1 promoter methylation and inability of hypomethylating drug 5-Aza-2'-deoxycytidine and HDAC inhibitor trichostatin A to reactivate FOXO1 expression suggest that loss of FOXO1 expression is due to mechanisms other than promoter methylation/acetylation. Inhibition of PI3K by LY294002 decreased the level of p-AKT1 and activated FOXO1 transcription factor. We demonstrate that activation of FOXO1 induces apoptosis, cell proliferation arrest, and decreased cell viability in cervical cancer cell lines. Our data suggest that frequent down regulation of FOXO1 and its functional inactivation may be due to post-translational modifications in cervical cancer. Together, these observations suggest that activation of FOXO1 and its nuclear sequestration is critical in the regulation of cell proliferation, cell viability and apoptosis in cervical cancer. Hence, PI3K/AKT pathway may be a potential molecular target for cervical cancer therapy.
Collapse
Affiliation(s)
- Shyam Babu Prasad
- 1. Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi-221 005, India
| | - Suresh Singh Yadav
- 1. Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi-221 005, India
| | - Mitali Das
- 1. Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi-221 005, India
| | - H B Govardhan
- 2. Department of Radiotherapy and Radiation Medicine, Banaras Hindu University, Varanasi-221 005, India
| | - Lakshmi Kant Pandey
- 3. Department of Obstetrics and Gynaecology, Banaras Hindu University, Varanasi-221 005, India
| | - Sunita Singh
- 4. Department of Zoology, Mahila Mahavidyalaya; Banaras Hindu University, Varanasi-221 005, India
| | - Satyajit Pradhan
- 2. Department of Radiotherapy and Radiation Medicine, Banaras Hindu University, Varanasi-221 005, India
| | - Gopeshwar Narayan
- 1. Cancer Genetics Laboratory, Department of Molecular and Human Genetics, Banaras Hindu University, Varanasi-221 005, India
| |
Collapse
|
33
|
Sousa V, Bastos B, Silva M, Alarcão AM, Carvalho L. WITHDRAWN: Bronchial-pulmonary adenocarcinoma subtyping relates with different molecular pathways. REVISTA PORTUGUESA DE PNEUMOLOGIA 2014:S0873-2159(14)00099-3. [PMID: 25131502 DOI: 10.1016/j.rppneu.2014.05.006] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/30/2013] [Revised: 05/20/2014] [Accepted: 05/30/2014] [Indexed: 10/24/2022] Open
Abstract
This article has been withdrawn for editorial reasons because the journal will be published only in English. In order to avoid duplicated records, this article can be found at http://dx.doi.org/10.1016/j.rppnen.2014.05.006. The Publisher apologizes for any inconvenience this may cause. The full Elsevier Policy on Article Withdrawal can be found at http://www.elsevier.com/locate/withdrawalpolicy.
Collapse
Affiliation(s)
- Vítor Sousa
- IAP-FMUC-Institute of Pathology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Pathology Institute, Coimbra University Hospital (HUC-CHUC), Coimbra, Portugal.
| | - Bruno Bastos
- IAP-FMUC-Institute of Pathology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Maria Silva
- IAP-FMUC-Institute of Pathology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Ana Maria Alarcão
- IAP-FMUC-Institute of Pathology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal
| | - Lina Carvalho
- IAP-FMUC-Institute of Pathology, Faculty of Medicine, University of Coimbra, Coimbra, Portugal; Pathology Institute, Coimbra University Hospital (HUC-CHUC), Coimbra, Portugal
| |
Collapse
|
34
|
Jane EP, Premkumar DR, Morales A, Foster KA, Pollack IF. Inhibition of phosphatidylinositol 3-kinase/AKT signaling by NVP-BKM120 promotes ABT-737-induced toxicity in a caspase-dependent manner through mitochondrial dysfunction and DNA damage response in established and primary cultured glioblastoma cells. J Pharmacol Exp Ther 2014; 350:22-35. [PMID: 24741074 PMCID: PMC4056270 DOI: 10.1124/jpet.114.212910] [Citation(s) in RCA: 31] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/08/2014] [Accepted: 04/14/2014] [Indexed: 12/16/2022] Open
Abstract
Identification of therapeutic strategies that might enhance the efficacy of B-cell lymphoma-2 (Bcl-2) inhibitor ABT-737 [N-{4-[4-(4-chloro-biphenyl-2-ylmethyl)-piperazin-1-yl]-benzoyl}-4-(3-dimethylamino-1-phenylsulfanylmethyl-propylamino)-3-nitro-benzenesulfonamide] is of great interest in many cancers, including glioma. Our recent study suggested that Akt is a crucial mediator of apoptosis sensitivity in response to ABT-737 in glioma cell lines. Inhibitors of phosphatidylinositol 3-kinase (PI3K)/Akt are currently being assessed clinically in patients with glioma. Because PI3K/Akt inhibition would be expected to have many proapoptotic effects, we hypothesized that there may be unique synergy between PI3K inhibitors and Bcl-2 homology 3 mimetics. Toward this end, we assessed the combination of the PI3K/Akt inhibitor NVP-BKM120 [5-(2,6-dimorpholinopyrimidin-4-yl)-4-(trifluoromethyl)pyridin-2-amine] and the Bcl-2 family inhibitor ABT-737 in established and primary cultured glioma cells. We found that the combined treatment with these agents led to a significant activation of caspase-8 and -3, PARP, and cell death, irrespective of PTEN status. The enhanced lethality observed with this combination also appears dependent on the loss of mitochondrial membrane potential and release of cytochrome c, smac/DIABLO, and apoptosis-inducing factor to the cytosol. Further study revealed that the upregulation of Noxa, truncation of Bid, and activation of Bax and Bak caused by these inhibitors were the key factors for the synergy. In addition, we demonstrated the release of proapoptotic proteins Bim and Bak from Mcl-1. We found defects in chromosome segregation leading to multinuclear cells and loss of colony-forming ability, suggesting the potential use of NVP-BKM120 as a promising agent to improve the anticancer activities of ABT-737.
Collapse
Affiliation(s)
- Esther P Jane
- Department of Neurologic Surgery, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania (E.P.J., D.R.P., K.A.F., I.F.P.); University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (E.P.J., D.R.P., A.M., K.A.F., I.F.P.); and University of Pittsburgh Brain Tumor Center, Pittsburgh, Pennsylvania (K.A.F., I.F.P.)
| | - Daniel R Premkumar
- Department of Neurologic Surgery, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania (E.P.J., D.R.P., K.A.F., I.F.P.); University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (E.P.J., D.R.P., A.M., K.A.F., I.F.P.); and University of Pittsburgh Brain Tumor Center, Pittsburgh, Pennsylvania (K.A.F., I.F.P.)
| | - Alejandro Morales
- Department of Neurologic Surgery, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania (E.P.J., D.R.P., K.A.F., I.F.P.); University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (E.P.J., D.R.P., A.M., K.A.F., I.F.P.); and University of Pittsburgh Brain Tumor Center, Pittsburgh, Pennsylvania (K.A.F., I.F.P.)
| | - Kimberly A Foster
- Department of Neurologic Surgery, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania (E.P.J., D.R.P., K.A.F., I.F.P.); University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (E.P.J., D.R.P., A.M., K.A.F., I.F.P.); and University of Pittsburgh Brain Tumor Center, Pittsburgh, Pennsylvania (K.A.F., I.F.P.)
| | - Ian F Pollack
- Department of Neurologic Surgery, Children's Hospital of Pittsburgh, Pittsburgh, Pennsylvania (E.P.J., D.R.P., K.A.F., I.F.P.); University of Pittsburgh, School of Medicine, Pittsburgh, Pennsylvania (E.P.J., D.R.P., A.M., K.A.F., I.F.P.); and University of Pittsburgh Brain Tumor Center, Pittsburgh, Pennsylvania (K.A.F., I.F.P.)
| |
Collapse
|
35
|
Fulda S. Synthetic lethality by co-targeting mitochondrial apoptosis and PI3K/Akt/mTOR signaling. Mitochondrion 2014; 19 Pt A:85-7. [PMID: 24780492 DOI: 10.1016/j.mito.2014.04.011] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/27/2014] [Revised: 04/16/2014] [Accepted: 04/18/2014] [Indexed: 11/29/2022]
Abstract
Small-molecule inhibitors that antagonize anti-apoptotic Bcl-2 proteins such as BH3 mimetics are currently considered as promising cancer therapeutics to engage the mitochondrial pathway of apoptosis in cancer cells. However, BH3 mimetics may be effective as monotherapy only in cancers that critically depend on anti-apoptotic Bcl-2 proteins for their survival. Since most cancers have evolved multiple strategies to evade programmed cell death, concomitant targeting of several signaling transduction pathways becomes more and more relevant. The current review highlights the potential of combined inhibition of anti-apoptotic Bcl-2 proteins together with the PI3K/Akt/mTOR signaling cascade to trigger apoptosis in cancer cells.
Collapse
Affiliation(s)
- Simone Fulda
- Institute for Experimental Cancer Research in Pediatrics, Goethe-University, Komturstr. 3a, 60528 Frankfurt, Germany.
| |
Collapse
|
36
|
Pareja F, Macleod D, Shu C, Crary JF, Canoll PD, Ross AH, Siegelin MD. PI3K and Bcl-2 inhibition primes glioblastoma cells to apoptosis through downregulation of Mcl-1 and Phospho-BAD. Mol Cancer Res 2014; 12:987-1001. [PMID: 24757258 DOI: 10.1158/1541-7786.mcr-13-0650] [Citation(s) in RCA: 68] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
UNLABELLED Glioblastoma multiforme (GBM) is a highly malignant human brain neoplasm with limited therapeutic options. GBMs display a deregulated apoptotic pathway with high levels of the antiapoptotic Bcl-2 family of proteins and overt activity of the phosphatidylinositol 3-kinase (PI3K) signaling pathway. Therefore, combined interference of the PI3K pathway and the Bcl-2 family of proteins is a reasonable therapeutic strategy. ABT-263 (Navitoclax), an orally available small-molecule Bcl-2 inhibitor, and GDC-0941, a PI3K inhibitor, were used to treat established glioblastoma and glioblastoma neurosphere cells, alone or in combination. Although GDC-0941 alone had a modest effect on cell viability, treatment with ABT-263 displayed a marked reduction of cell viability and induction of apoptotic cell death. Moreover, combinatorial therapy using ABT-263 and GDC-0941 showed an enhanced effect, with a further decrease in cellular viability. Furthermore, combination treatment abrogated the ability of stem cell-like glioma cells to form neurospheres. ABT-263 and GDC-0941, in combination, resulted in a consistent and significant increase of Annexin V positive cells and loss of mitochondrial membrane potential compared with either monotherapy. The combination treatment led to enhanced cleavage of both initiator and effector caspases. Mechanistically, GDC-0941 depleted pAKT (Serine 473) levels and suppressed Mcl-1 protein levels, lowering the threshold for the cytotoxic actions of ABT-263. GDC-0941 decreased Mcl-1 in a posttranslational manner and significantly decreased the half-life of Mcl-1 protein. Ectopic expression of human Mcl-1 mitigated apoptotic cell death induced by the drug combination. Furthermore, GDC-0941 modulated the phosphorylation status of BAD, thereby further enhancing ABT-263-mediated cell death. IMPLICATIONS Combination therapy with ABT-263 and GDC-0941 has novel therapeutic potential by specifically targeting aberrantly active, deregulated pathways in GBM, overcoming endogenous resistance to apoptosis.
Collapse
Affiliation(s)
- Fresia Pareja
- Authors' Affiliations: Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York; and
| | - David Macleod
- Authors' Affiliations: Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York; and
| | - Chang Shu
- Authors' Affiliations: Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York; and
| | - John F Crary
- Authors' Affiliations: Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York; and
| | - Peter D Canoll
- Authors' Affiliations: Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York; and
| | - Alonzo H Ross
- Department of Biochemistry and Molecular Pharmacology, University of Massachusetts Medical School, Worcester, Massachusetts
| | - Markus D Siegelin
- Authors' Affiliations: Department of Pathology & Cell Biology, Columbia University Medical Center, New York, New York; and
| |
Collapse
|
37
|
Beech RD, Leffert JJ, Lin A, Sylvia LG, Umlauf S, Mane S, Zhao H, Bowden C, Calabrese JR, Friedman ES, Ketter TA, Iosifescu DV, Reilly-Harrington NA, Ostacher M, Thase ME, Nierenberg A. Gene-expression differences in peripheral blood between lithium responders and non-responders in the Lithium Treatment-Moderate dose Use Study (LiTMUS). THE PHARMACOGENOMICS JOURNAL 2014; 14:182-91. [PMID: 23670706 DOI: 10.1038/tpj.2013.16] [Citation(s) in RCA: 25] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/13/2012] [Revised: 02/15/2013] [Accepted: 03/18/2013] [Indexed: 11/08/2022]
Abstract
This study was designed to identify genes whose expression in peripheral blood may serve as early markers for treatment response to lithium (Li) in patients with bipolar disorder. Although changes in peripheral blood gene-expression may not relate directly to mood symptoms, differences in treatment response at the biochemical level may underlie some of the heterogeneity in clinical response to Li. Subjects were randomized to treatment with (n=28) or without (n=32) Li. Peripheral blood gene-expression was measured before and 1 month after treatment initiation, and treatment response was assessed after 6 months. In subjects treated with Li, 62 genes were differentially regulated in treatment responders and non-responders. Of these, BCL2L1 showed the greatest difference between Li responders and non-responders. These changes were specific to Li responders (n=9), and were not seen in Li non-responders or patients treated without Li, suggesting that they may have specific roles in treatment response to Li.
Collapse
Affiliation(s)
- R D Beech
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - J J Leffert
- Department of Psychiatry, Yale University School of Medicine, New Haven, CT, USA
| | - A Lin
- Keck Foundation Biotechnology Biostatistics Resource, Yale University School of Medicine, New Haven, CT, USA
| | - L G Sylvia
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| | - S Umlauf
- Center for Genome Analysis, Yale University School of Medicine, New Haven, CT, USA
| | - S Mane
- Center for Genome Analysis, Yale University School of Medicine, New Haven, CT, USA
| | - H Zhao
- Department of Epidemiology and Public Health, Yale University School of Medicine, New Haven, CT, USA
| | - C Bowden
- Departments of Psychiatry and Pharmacology, University of Texas Health Science, San Antonio, TX, USA
| | - J R Calabrese
- Department of Psychiatry, Case Western Reserve University, Cleveland, OH, USA
| | - E S Friedman
- Department of Psychiatry, University of Pittsburgh Medical Center, Pittsburgh, PA, USA
| | - T A Ketter
- Department of Psychiatry and Behavioral Science, Stanford University School of Medicine, Stanford, CA, USA
| | - D V Iosifescu
- Departments of Psychiatry and Neuroscience, Mount Sinai Medical Center, New York, NY, USA
| | | | - M Ostacher
- Department of Psychiatry and Behavioral Science, Stanford University School of Medicine, Stanford, CA, USA
| | - M E Thase
- Department of Psychiatry, University of Pennsylvania, Philadelphia, PA, USA
| | - A Nierenberg
- Department of Psychiatry, Massachusetts General Hospital, Boston, MA, USA
| |
Collapse
|
38
|
Jebahi A, Villedieu M, Pétigny-Lechartier C, Brotin E, Louis MH, Abeilard E, Giffard F, Guercio M, Briand M, Gauduchon P, Lheureux S, Poulain L. PI3K/mTOR dual inhibitor NVP-BEZ235 decreases Mcl-1 expression and sensitizes ovarian carcinoma cells to Bcl-xL-targeting strategies, provided that Bim expression is induced. Cancer Lett 2014; 348:38-49. [PMID: 24650799 DOI: 10.1016/j.canlet.2014.03.001] [Citation(s) in RCA: 44] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2013] [Revised: 02/10/2014] [Accepted: 03/04/2014] [Indexed: 01/25/2023]
Abstract
We previously showed that Bcl-xL and Mcl-1 cooperatively protect platinum-resistant ovarian cancer cells from apoptosis. Here we assessed the anticancer potential of combining ABT-737-induced inhibition of Bcl-xL with Mcl-1 inhibition via PI3K/Akt/mTOR pathway disruption using NVP-BEZ235. NVP-BEZ235 inhibited cell proliferation without inducing apoptosis. It strongly repressed Mcl-1 expression and induced Puma expression in both cell lines tested while differentially modulating Bim between the two. Interestingly, NVP-BEZ235 efficiently sensitized ovarian carcinoma cells to ABT-737, provided that Bim expression was induced. Moreover, inhibiting the ERK1/2 pathway restored Bim expression and sensitized low Bim-expressing cancer cells to the BEZ235/ABT-737 treatment.
Collapse
Affiliation(s)
- Abdelghani Jebahi
- Normandy University, France; UNICAEN, "Biology and Innovative Therapeutics of Locally Aggressive Cancers" Unit (EA 4656), Caen, France; François Baclesse Comprehensive Cancer Centre, Caen, France
| | - Marie Villedieu
- Normandy University, France; UNICAEN, "Biology and Innovative Therapeutics of Locally Aggressive Cancers" Unit (EA 4656), Caen, France; François Baclesse Comprehensive Cancer Centre, Caen, France; (d)On secondment from ISPB, Faculte de Pharmacie, Universite Lyon 1, Lyon, France.
| | - Cécile Pétigny-Lechartier
- Normandy University, France; UNICAEN, "Biology and Innovative Therapeutics of Locally Aggressive Cancers" Unit (EA 4656), Caen, France; François Baclesse Comprehensive Cancer Centre, Caen, France
| | - Emilie Brotin
- Normandy University, France; UNICAEN, "Biology and Innovative Therapeutics of Locally Aggressive Cancers" Unit (EA 4656), Caen, France
| | - Marie-Hélène Louis
- Normandy University, France; UNICAEN, "Biology and Innovative Therapeutics of Locally Aggressive Cancers" Unit (EA 4656), Caen, France; François Baclesse Comprehensive Cancer Centre, Caen, France
| | - Edwige Abeilard
- Normandy University, France; UNICAEN, "Biology and Innovative Therapeutics of Locally Aggressive Cancers" Unit (EA 4656), Caen, France; François Baclesse Comprehensive Cancer Centre, Caen, France
| | - Florence Giffard
- Normandy University, France; UNICAEN, "Biology and Innovative Therapeutics of Locally Aggressive Cancers" Unit (EA 4656), Caen, France; François Baclesse Comprehensive Cancer Centre, Caen, France
| | - Marika Guercio
- Normandy University, France; UNICAEN, "Biology and Innovative Therapeutics of Locally Aggressive Cancers" Unit (EA 4656), Caen, France
| | - Mélanie Briand
- Normandy University, France; UNICAEN, "Biology and Innovative Therapeutics of Locally Aggressive Cancers" Unit (EA 4656), Caen, France; François Baclesse Comprehensive Cancer Centre, Caen, France
| | - Pascal Gauduchon
- Normandy University, France; UNICAEN, "Biology and Innovative Therapeutics of Locally Aggressive Cancers" Unit (EA 4656), Caen, France; François Baclesse Comprehensive Cancer Centre, Caen, France
| | - Stéphanie Lheureux
- Normandy University, France; UNICAEN, "Biology and Innovative Therapeutics of Locally Aggressive Cancers" Unit (EA 4656), Caen, France; François Baclesse Comprehensive Cancer Centre, Caen, France; Clinical Research Department, François Baclesse Comprehensive Cancer Centre, Caen, France; Oncologic Uro-Gynaecology Department, François Baclesse Comprehensive Cancer Centre, Caen, France
| | - Laurent Poulain
- Normandy University, France; UNICAEN, "Biology and Innovative Therapeutics of Locally Aggressive Cancers" Unit (EA 4656), Caen, France; François Baclesse Comprehensive Cancer Centre, Caen, France
| |
Collapse
|
39
|
Secreto F, Manske M, Price-Troska T, Ziesmer S, Hodge LS, Ansell SM, Cerhan JR, Novak AJ. B-cell activating factor-receptor specific activation of tumor necrosis factor receptor associated factor 6 and the phosphatidyl inositol 3-kinase pathway in lymphoma B cells. Leuk Lymphoma 2014; 55:1884-92. [PMID: 24206092 DOI: 10.3109/10428194.2013.862619] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
B-cell activating factor-receptor (BAFF-R) is the primary BAFF receptor that is responsible for promoting B-cell development and survival. Malignant B-cells exploit the BAFF/BAFF-R system, and high serum BAFF levels or genetic alterations in BAFF receptors have been found in B-cell cancers. BAFF signaling impacts pro-survival pathways. However, other than nuclear factor-κB2 (NF-κB2), little is known about the specific pathways activated by individual BAFF receptors. Using a novel BAFF-R expression model we have demonstrated that activation of BAFF-R, independent of transmembrane activator and cytophilin ligand interactor (TACI) and B-cell maturation antigen (BCMA), can induce phosphorylation of Akt and glycogen synthase kinase 3β (GSK3β). Expression of an activated form of BAFF-R also enhanced a pro-survival gene expression pattern, including the novel BAFF-regulated gene Pin1, whose expression was phosphatidyl inositol 3-kinase (PI3K)-dependent. Additionally, we showed that TRAF6 is essential for mediating BAFF-R dependent activation of Akt. Together these data describe a novel role for TRAF6 in BAFF-R-specific activation of the PI3K pathway and provide evidence suggesting a new role for Pin1 in BAFF-R signaling.
Collapse
Affiliation(s)
- Frank Secreto
- Division of Hematology, Mayo Clinic , Rochester, MN , USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Potter DS, Kelly P, Denneny O, Juvin V, Stephens LR, Dive C, Morrow CJ. BMX acts downstream of PI3K to promote colorectal cancer cell survival and pathway inhibition sensitizes to the BH3 mimetic ABT-737. Neoplasia 2014; 16:147-57. [PMID: 24709422 PMCID: PMC3978395 DOI: 10.1593/neo.131376] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2013] [Revised: 12/12/2013] [Accepted: 01/17/2014] [Indexed: 01/10/2023]
Abstract
Evasion of apoptosis is a hallmark of cancer, and reversing this process by inhibition of survival signaling pathways is a potential therapeutic strategy. Phosphoinositide 3-kinase (PI3K) signaling can promote cell survival and is upregulated in solid tumor types, including colorectal cancer (CRC), although these effects are context dependent. The role of PI3K in tumorigenesis combined with their amenability to specific inhibition makes them attractive drug targets. However, we observed that inhibition of PI3K in HCT116, DLD-1, and SW620 CRC cells did not induce apoptotic cell death. Moreover, these cells were relatively resistant to the Bcl-2 homology domain 3 (BH3) mimetic ABT-737, which directly targets the Bcl-2 family of apoptosis regulators. To test the hypothesis that PI3K inhibition lowers the apoptotic threshold without causing apoptosis per se, PI3K inhibitors were combined with ABT-737. PI3K inhibition enhanced ABT-737-induced apoptosis by 2.3- to 4.5-fold and reduced expression levels of MCL-1, the resistance biomarker for ABT-737. PI3K inhibition enhanced ABT-737-induced apoptosis a further 1.4- to 2.4-fold in CRC cells with small interfering RNA-depleted MCL-1, indicative of additional sensitizing mechanisms. The observation that ABT-737-induced apoptosis was unaffected by inhibition of PI3K downstream effectors AKT and mTOR, implicated a novel PI3K-dependant pathway. To elucidate this, an RNA interference (RNAi) screen of potential downstream effectors of PI3K signaling was conducted, which demonstrated that knockdown of the TEC kinase BMX sensitized to ABT-737. This suggests that BMX is an antiapoptotic downstream effector of PI3K, independent of AKT.
Collapse
Affiliation(s)
- Danielle S Potter
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Paul Kelly
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Olive Denneny
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom
| | - Veronique Juvin
- Inositide Laboratory, Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Len R Stephens
- Inositide Laboratory, Babraham Institute, Babraham Research Campus, Cambridge, United Kingdom
| | - Caroline Dive
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom.
| | - Christopher J Morrow
- Clinical and Experimental Pharmacology Group, Cancer Research UK Manchester Institute, University of Manchester, Manchester, United Kingdom.
| |
Collapse
|
41
|
Shao H, Jing K, Mahmoud E, Huang H, Fang X, Yu C. Apigenin sensitizes colon cancer cells to antitumor activity of ABT-263. Mol Cancer Ther 2013; 12:2640-50. [PMID: 24126433 DOI: 10.1158/1535-7163.mct-13-0066] [Citation(s) in RCA: 82] [Impact Index Per Article: 6.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Apigenin is an edible plant-derived flavonoid that shows modest antitumor activities in vitro and in vivo. Apigenin treatment resulted in cell growth arrest and apoptosis in various types of tumors by modulating several signaling pathways. In the present study, we evaluated interactions between apigenin and ABT-263 in colon cancer cells. We observed a synergistic effect between apigenin and ABT-263 on apoptosis of colon cancer cells. ABT-263 alone induced limited cell death while upregulating expression of Mcl-1, a potential mechanism for the acquired resistance to ABT-263. The presence of apigenin antagonized ABT-263-induced Mcl-1 upregulation and dramatically enhanced ABT-263-induced cell death. Meanwhile, apigenin suppressed AKT and ERK activation. Inactivation of either AKT or ERK by lentivirus-transduced shRNA or treatment with specific small-molecule inhibitors of these pathways enhanced ABT-263-induced cell death, mirroring the effect of apigenin. Moreover, the combination response was associated with upregulation of Bim and activation of Bax. Downregulation of Bax eliminated the synergistic effect of apigenin and ABT-263 on cell death. Xenograft studies in SCID mice showed that the combined treatment with apigenin and ABT-263 inhibited tumor growth by up to 70% without obvious adverse effects, while either agent only inhibited around 30%. Our results demonstrate a novel strategy to enhance ABT-263-induced antitumor activity in human colon cancer cells by apigenin via inhibition of the Mcl-1, AKT, and ERK prosurvival regulators.
Collapse
Affiliation(s)
- Huanjie Shao
- Corresponding Authors: Huanjie Shao, Department of Biochemistry and Molecular Biology, Virginia Commonwealth University School of Medicine, 1101 East Marshall Street, Richmond, VA 23298.
| | | | | | | | | | | |
Collapse
|
42
|
Sale MJ, Cook SJ. That which does not kill me makes me stronger; combining ERK1/2 pathway inhibitors and BH3 mimetics to kill tumour cells and prevent acquired resistance. Br J Pharmacol 2013; 169:1708-22. [PMID: 23647573 PMCID: PMC3753831 DOI: 10.1111/bph.12220] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/24/2013] [Revised: 04/10/2013] [Accepted: 04/25/2013] [Indexed: 01/07/2023] Open
Abstract
UNLABELLED Oncogenic mutations in RAS or BRAF can drive the inappropriate activation of the ERK1/2. In many cases, tumour cells adapt to become addicted to this deregulated ERK1/2 signalling for their proliferation, providing a therapeutic window for tumour-selective growth inhibition. As a result, inhibition of ERK1/2 signalling by BRAF or MEK1/2 inhibitors is an attractive therapeutic strategy. Indeed, the first BRAF inhibitor, vemurafenib, has now been approved for clinical use, while clinical evaluation of MEK1/2 inhibitors is at an advanced stage. Despite this progress, it is apparent that tumour cells adapt quickly to these new targeted agents so that tumours with acquired resistance can emerge within 6-9 months of primary treatment. One of the major reasons for this is that tumour cells typically respond to BRAF or MEK1/2 inhibitors by undergoing a G1 cell cycle arrest rather than dying. Indeed, although inhibition of ERK1/2 invariably increases the expression of pro-apoptotic BCL2 family proteins, tumour cells undergo minimal apoptosis. This cytostatic response may simply provide the cell with the opportunity to adapt and acquire resistance. Here we discuss recent studies that demonstrate that combination of BRAF or MEK1/2 inhibitors with inhibitors of pro-survival BCL2 proteins is synthetic lethal for ERK1/2-addicted tumour cells. This combination effectively transforms the cytostatic response of BRAF and MEK1/2 inhibitors into a striking apoptotic cell death response. This not only augments the primary efficacy of BRAF and MEK1/2 inhibitors but delays the onset of acquired resistance to these agents, validating their combination in the clinic. LINKED ARTICLES This article is part of a themed section on Emerging Therapeutic Aspects in Oncology. To view the other articles in this section visit http://dx.doi.org/10.1111/bph.2013.169.issue-8.
Collapse
Affiliation(s)
- Matthew J Sale
- Signalling Laboratory, The Babraham Institute, Babraham Research Campus, Cambridge, UK.
| | | |
Collapse
|
43
|
Wang J, Qian J, Hoeksema MD, Zou Y, Espinosa AV, Rahman SMJ, Zhang B, Massion PP. Integrative genomics analysis identifies candidate drivers at 3q26-29 amplicon in squamous cell carcinoma of the lung. Clin Cancer Res 2013; 19:5580-90. [PMID: 23908357 DOI: 10.1158/1078-0432.ccr-13-0594] [Citation(s) in RCA: 57] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/05/2023]
Abstract
PURPOSE Chromosome 3q26-29 is a critical region of genomic amplification in lung squamous cell carcinomas (SCC). Identification of candidate drivers in this region could help uncover new mechanisms in the pathogenesis and potentially new targets in SCC of the lung. EXPERIMENTAL DESIGN We conducted a meta-analysis of seven independent datasets containing a total of 593 human primary SCC samples to identify consensus candidate drivers in 3q26-29 amplicon. Through integrating protein-protein interaction network information, we further filtered for candidates that may function together in a network. Computationally predicted candidates were validated using RNA interference (RNAi) knockdown and cell viability assays. Clinical relevance of the experimentally supported drivers was evaluated in an independent cohort of 52 lung SCC patients using survival analysis. RESULTS The meta-analysis identified 20 consensus candidates, among which four (SENP2, DCUN1D1, DVL3, and UBXN7) are involved in a small protein-protein interaction network. Knocking down any of the four proteins led to cell growth inhibition of the 3q26-29-amplified SCC. Moreover, knocking down of SENP2 resulted in the most significant cell growth inhibition and downregulation of DCUN1D1 and DVL3. Importantly, a gene expression signature composed of SENP2, DCUN1D1, and DVL3 stratified patients into subgroups with different response to adjuvant chemotherapy. CONCLUSION Together, our findings show that SENP2, DCUN1D1, and DVL3 are candidate driver genes in the 3q26-29 amplicon of SCC, providing novel insights into the molecular mechanisms of disease progression and may have significant implication in the management of SCC of the lung.
Collapse
Affiliation(s)
- Jing Wang
- Authors' Affiliations: Department of Biomedical Informatics, Division of Allergy, Pulmonary and Critical Care Medicine, Vanderbilt University School of Medicine; and Veterans Affairs, Tennessee Valley Health Care Systems, Nashville, Tennessee
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Fang Z, Tang Y, Jiao W, Xing Z, Guo Z, Wang W, Shi B, Xu Z, Liu Z. Nitidine chloride inhibits renal cancer cell metastasis via suppressing AKT signaling pathway. Food Chem Toxicol 2013; 60:246-51. [PMID: 23911800 DOI: 10.1016/j.fct.2013.07.062] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2013] [Revised: 07/20/2013] [Accepted: 07/22/2013] [Indexed: 10/26/2022]
Abstract
Nitidine Chloride (NC) has been shown to have anti-cancer effects on various tumors. However, whether NC could exert anti-metastasis activity in renal cancer cells and the underlying mechanisms have not been elucidated. In this work, our data demonstrated the anti-metastasis effects of NC on renal cancer cells in vitro. With scratch assay and transwell assays, we found that NC potently suppressed the migration and invasion of 786-O and A498 cells. Mechanistically, we presented that NC significantly decreased phosphorylation of AKT, accompanied by down-regulation of MMP-2 and MMP-9. Furthermore, a specific AKT inhibitor, LY294002, could enhance the anti-metastasis effects of NC, which indicated that NC suppressed metastasis of renal cancer cells partly via inhibition of AKT activity. Taken together, our results imply that NC can be developed as a potential anti-metastasis agent to renal cancer.
Collapse
Affiliation(s)
- Zhiqing Fang
- Department of Urology, Qilu Hospital of Shandong University, Ji'nan, Shandong, China; The Key Laboratory of Cardiovascular Remodeling and Function Research, Chinese Ministry of Education and Chinese Ministry of Public Health, Qilu Hospital of Shandong University, Ji'nan, Shandong, China
| | | | | | | | | | | | | | | | | |
Collapse
|
45
|
Bhattacharyya S, Pal PB, Sil PC. A 35 kD Phyllanthus niruri protein modulates iron mediated oxidative impairment to hepatocytes via the inhibition of ERKs, p38 MAPKs and activation of PI3k/Akt pathway. Food Chem Toxicol 2013; 56:119-130. [PMID: 23435124 DOI: 10.1016/j.fct.2013.02.013] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/17/2012] [Revised: 02/01/2013] [Accepted: 02/09/2013] [Indexed: 12/21/2022]
Abstract
It has been reported that the herb, Phyllanthus niruri, possess antioxidant, anti-infection, anti-asthmatic, anti-diuretic, anti-soresis and many more beneficial activities. The goal of our present study was to evaluate the protective role of a 35 kD protein (PNP) isolated from this herb against iron-induced cytotoxicity in murine hepatocytes. Exposure of hepatocytes to iron (FeSO4) caused elevation of reactive oxygen species (ROS) production, enhanced lipid peroxidation and protein carbonylation, depleted glutathione levels, decreased the antioxidant power (FRAP) of the cells and reduced cell viability. Iron mediated cytotoxicity disrupted mitochondrial membrane potential (Δψm) and thereby caused apoptosis mainly by the intrinsic pathway via the down-regulation of IκBα with a concomitant up-regulation of NF-kB as well as the phosphorylation of ERKs and p38 MAP kinases. In addition, iron-induced cytotoxicity disrupted the normal balance of Bcl-2 family proteins in hepatocytes. Incubation of hepatocytes with PNP, however, protected the cells from apoptosis by stabilizing the mitochondria and arresting the release of cytochrome c. It also suppressed caspase activation and cleavage of PARP. Moreover, this protein has strong free radical scavenging activity and thereby scavenged ROS extensively. Combining all, results suggest that simultaneous treatment with PNP might suppress the iron-induced cytotoxicity in hepatocytes.
Collapse
Affiliation(s)
- Sudip Bhattacharyya
- Division of Molecular Medicine, Bose Institute, P-1/12, CIT Scheme VII M, Kolkata 700 054, West Bengal, India
| | | | | |
Collapse
|
46
|
Zhu S, Cohen MB, Bjorge JD, Mier JW, Cho DC. PI3K inhibition potentiates Bcl-2-dependent apoptosis in renal carcinoma cells. J Cell Mol Med 2013; 17:377-85. [PMID: 23387989 PMCID: PMC3612143 DOI: 10.1111/jcmm.12019] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/17/2012] [Accepted: 12/28/2012] [Indexed: 01/31/2023] Open
Abstract
Inhibitors of PI3-K/Akt are currently being assessed clinically in patients with advanced RCC. Identification of therapeutic strategies that might enhance the efficacy of PI3-K/Akt inhibitors is therefore of great interest. As PI3-K inhibition would be expected to have many pro-apoptotic effects, we hypothesized that there may be unique synergy between PI3-K inhibitors and BH3-mimetics. Towards this end, we assessed the combination of the PI3K inhibitor LY 294002 and the Bcl-2 family inhibitor ABT-737 in RCC cell lines. We found that the combinatorial treatment with these agents led to a significant increase in PARP cleavage and cell death in all RCC cell lines. The synergized cell death was correlated with decreased levels of Mcl-1 and XIAP, and increased levels in Bim, and appears critically dependent upon the activation of caspase 3 and 8. The enhanced lethality observed with the combination also appears dependent upon the regulation of XIAP, Mcl-1 and Bim levels. Our results suggest that the combination of PI3-K inhibitors with BH3-mimetics may be a viable therapeutic strategy in RCC.
Collapse
Affiliation(s)
- Shudong Zhu
- Division of Hematology and Oncology, Beth Israel Deaconess Medical Center, Harvard Medical School, Boston, MA 02215, USA.
| | | | | | | | | |
Collapse
|
47
|
Guo R, Wang Y, Shi WY, Liu B, Hou SQ, Liu L. MicroRNA miR-491-5p targeting both TP53 and Bcl-XL induces cell apoptosis in SW1990 pancreatic cancer cells through mitochondria mediated pathway. Molecules 2012; 17:14733-47. [PMID: 23519249 PMCID: PMC6268397 DOI: 10.3390/molecules171214733] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/20/2012] [Revised: 12/03/2012] [Accepted: 12/06/2012] [Indexed: 11/24/2022] Open
Abstract
MicroRNA (miRNA) actively participates in a broad range of cellular processes such as proliferation, differentiation, cell survival and apoptosis. Deregulated expression of miRNA may affect cell growth and eventually lead to cancer. In this study, we found that hsa-miR491-5p (miR491-5p) displays a significantly high level of expression in normal human pancreas tissue versus pancreatic cancer cells. Targeted site prediction indicated that both Bcl-XL and TP53 contain miR-491-5p recognizing sites in their 3' UTRs. Overexpression of miR-491-5p in the pancreatic cancer cell line SW1990 effectively inhibited both endogenous Bcl-XL and TP53 gene expressions. Mutagenesis at the seed match region of both targeted genes further confirmed the specificity of miR491-5p recognition. Cell proliferation rate was inversely related to the increased doses of miR-491-5p. Flow cytometric analysis showed that the proportions of total apoptotic and early apoptotic cells were significantly induced as the dose of miR491-5p increased. Moreover, a mechanistic study indicated that miR-R491-5p-mediated cell apoptosis was associated with the activation of intrinsic mitochondria mediated pathways. miR491-5p also markedly inhibited mitogenic signaling pathways such as STAT3 and PI-3K/Akt, but not Ras/MAPK. Thus, our results demonstrated that miR491-5p could effectively target both Bcl-xL and TP53 and induce cell apoptosis independent of TP53.
Collapse
Affiliation(s)
| | | | | | | | | | - Li Liu
- Author to whom correspondence should be addressed; E-Mail: ; Tel.: +86-10-6915-6454; Fax: +86-10-6523-3768
| |
Collapse
|
48
|
He X, Wang Y, Hu H, Zhang Z. In vitro and in vivo antimammary tumor activities and mechanisms of the apple total triterpenoids. JOURNAL OF AGRICULTURAL AND FOOD CHEMISTRY 2012; 60:9430-6. [PMID: 22924395 DOI: 10.1021/jf3026925] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/01/2023]
Abstract
Consumption of apples has been linked to the prevention of various chronic diseases, including tumors and cardiovascular diseases. The apple total triterpenoid content (ATT) was extracted and concentrated from apple peels. The in vitro and in vivo antitumor activities, related antitumor mechanisms, were investigated. In vitro, ATT showed potent antiproliferative activities against human breast cancer (MCF-7, MDA-MB-231), human colon cancer (Caco-2), and human liver cancer (HepG2) cell lines. In vivo antitumor experiments showed that ATT could substantially reduce the occurrence and growth of mammary tumor with a good dose-dependent manner in a rat model. During the apoptosis in MDA-MB-231 cells induced by ATT, the caspase-independent pathway was involved in the process of apoptosis, and the mitochondrial transmembrane potential was markedly reduced. Also the PI3K/Akt/NF-κB pathway was activated. These results indicated that ATT-induced apoptosis of MDA-MB-231 cells may involve a mitochondrial-related pathway.
Collapse
Affiliation(s)
- Xiangjiu He
- School of Pharmaceutical Sciences, Wuhan University, Wuhan, China
| | | | | | | |
Collapse
|
49
|
Stamelos VA, Redman CW, Richardson A. Understanding sensitivity to BH3 mimetics: ABT-737 as a case study to foresee the complexities of personalized medicine. J Mol Signal 2012; 7:12. [PMID: 22898329 PMCID: PMC3477050 DOI: 10.1186/1750-2187-7-12] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/23/2012] [Accepted: 07/29/2012] [Indexed: 12/17/2022] Open
Abstract
BH3 mimetics such as ABT-737 and navitoclax bind to the BCL-2 family of proteins and induce apoptosis through the intrinsic apoptosis pathway. There is considerable variability in the sensitivity of different cells to these drugs. Understanding the molecular basis of this variability will help to determine which patients will benefit from these drugs. Furthermore, this understanding aids in the design of rational strategies to increase the sensitivity of cells which are otherwise resistant to BH3 mimetics. We discuss how the expression of BCL-2 family proteins regulates the sensitivity to ABT-737. One of these, MCL-1, has been widely described as contributing to resistance to ABT-737 which might suggest a poor response in patients with cancers that express levels of MCL-1. In some cases, resistance to ABT-737 conferred by MCL-1 is overcome by the expression of pro-apoptotic proteins that bind to apoptosis inhibitors such as MCL-1. However, the distribution of the pro-apoptotic proteins amongst the various apoptosis inhibitors also influences sensitivity to ABT-737. Furthermore, the expression of both pro- and anti-apoptotic proteins can change dynamically in response to exposure to ABT-737. Thus, there is significant complexity associated with predicting response to ABT-737. This provides a paradigm for the multiplicity of intricate factors that determine drug sensitivity which must be considered for the full implementation of personalized medicine.
Collapse
Affiliation(s)
- Vasileios A Stamelos
- Institute for Science and Technology in Medicine & School of Pharmacy, Guy Hilton Research Centre, Keele University, Thornburrow Drive, Stoke-on-Trent, Keele, ST4 7QB, UK.
| | | | | |
Collapse
|
50
|
Minai-Tehrani A, Park YC, Hwang SK, Kwon JT, Chang SH, Park SJ, Yu KN, Kim JE, Shin JY, Kim JH, Kang B, Hong SH, Cho MH. Aerosol delivery of kinase-deficient Akt1 attenuates Clara cell injury induced by naphthalene in the lungs of dual luciferase mice. J Vet Sci 2012; 12:309-17. [PMID: 22122896 PMCID: PMC3232389 DOI: 10.4142/jvs.2011.12.4.309] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/08/2023] Open
Abstract
Conventional lung cancer therapies are associated with poor survival rates; therefore, new approaches such as gene therapy are required for treating cancer. Gene therapies for treating lung cancer patients can involve several approaches. Among these, aerosol gene delivery is a potentially more effective approach. In this study, Akt1 kinase-deficient (KD) and wild-type (WT) Akt1 were delivered to the lungs of CMV-LucR-cMyc-IRES-LucF dual reporter mice through a nose only inhalation system using glucosylated polyethylenimine and naphthalene was administrated to the mice via intraperitoneal injection. Aerosol delivery of Akt1 WT and naphthalene treatment increased protein levels of downstream substrates of Akt signaling pathway while aerosol delivery of Akt1 KD did not. Our results showed that naphthalene affected extracellular signal-regulated kinase (ERK) protein levels, ERK-related signaling, and induced Clara cell injury. However, Clara cell injury induced by naphthalene was considerably attenuated in mice exposed to Akt1 KD. Furthermore, a dual luciferase activity assay showed that aerosol delivery of Akt1 WT and naphthalene treatment enhanced cap-dependent protein translation, while reduced cap-dependent protein translation was observed after delivering Akt1 KD. These studies demonstrated that our aerosol delivery is compatible for in vivo gene delivery.
Collapse
Affiliation(s)
- Arash Minai-Tehrani
- Laboratory of Toxicology, College of Veterinary Medicine, Seoul National University, Seoul 151-742, Korea
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|