1
|
Zhou Z, Sicairos B, Zhou J, Du Y. Proteomic Analysis Reveals Major Proteins and Pathways That Mediate the Effect of 17-β-Estradiol in Cell Division and Apoptosis in Breast Cancer MCF7 Cells. J Proteome Res 2024; 23:4835-4848. [PMID: 39392593 PMCID: PMC11536429 DOI: 10.1021/acs.jproteome.4c00102] [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: 02/13/2024] [Revised: 09/28/2024] [Accepted: 10/01/2024] [Indexed: 10/12/2024]
Abstract
Despite extensive research, the genes/proteins and pathways responsible for the physiological effects of estrogen remain elusive. In this study, we determined the effect of estrogen on global protein expression in breast cancer MCF7 cells using a proteomic method. The expression of 77 cytosolic, 74 nuclear, and 81 membrane/organelle proteins was significantly altered by 17-β-estradiol (E2). Protein enrichment analyses suggest that E2 may stimulate cell division primarily by promoting the G1 to S phase transition and advancing the G2/M checkpoint. The effect of E2 on cell survival was complex, as it could simultaneously enhance and inhibit apoptosis. Bioinformatics analysis suggests that E2 may enhance apoptosis by promoting the accumulation of the pore-forming protein Bax in the mitochondria and inhibit apoptosis by activating the PI3K/AKT/mTOR signaling pathway. We verified the activation of the PI3K signaling and the accumulation of Bax in the membrane/organelle fraction in E2-treated cells using immunoblotting. Treatment of MCF7 cells with E2 and the PI3K inhibitor Ly294002 significantly enhanced apoptosis compared to those treated with E2 alone, suggesting that combining estrogen with a PI3K inhibitor could be a promising strategy for treating ERα-positive breast cancer. Interestingly, many of the E2-upregulated proteins contained the HEAT, KH, and RRM domains.
Collapse
Affiliation(s)
| | | | | | - Yuchun Du
- Department of Biological
Sciences, University of Arkansas, Fayetteville, Arkansas 72701, United States
| |
Collapse
|
2
|
Yadav P, Bandyopadhyay A, Sarkar K. Enhancement of gold-curcumin nanoparticle mediated radiation response for improved therapy in cervical cancer: a computational approach and predictive pathway analysis. DISCOVER NANO 2024; 19:153. [PMID: 39292302 PMCID: PMC11410751 DOI: 10.1186/s11671-024-04104-7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 03/27/2024] [Accepted: 09/05/2024] [Indexed: 09/19/2024]
Abstract
Radiotherapy is prevalently applied for highly effective cancer therapy while the low specificity of radiation is deleterious to the nearby healthy cells. High-Z-based nanomaterials offer excellent radio-enhancement properties while natural products provide radioprotection. Modulation of the radiotherapeutic index via applying nanomaterials is feasible for effective treatment however, the scenario changes when simultaneous protection of non-cancerous cells is required. Here, we report the modulatory radiotherapeutic effect of curcumin conjugated gold nanoparticles in a single nanoformulation to pave the long-awaited hope of a single combination-based, cell-selective radio enhancer, and protectant for cancer radiotherapy. We have validated the effective radiation dose along with the combination of the radio-nano-modulator by a reverse experimentation statistical model. The concept was supported by different sets of experiments, like quantification of ROS generation, cell cycle monitoring, mitochondrial membrane potential measurement, etc. along with gene expression study, and predictive modeling of molecular pathways of the killing mechanism. In conclusion, the nanoconjugate showed a promise to become a candidate for the pH-dependent cell-specific radio-modulator.
Collapse
Affiliation(s)
- Priya Yadav
- Department of Microbiology, University of Kalyani, Kalyani, West Bengal, 741235, India
- Department of Biochemistry and Biotechnology, Annamalai University, Annamalai Nagar, Tamil Nadu, 608002, India
| | - Arghya Bandyopadhyay
- Department of Microbiology, University of Kalyani, Kalyani, West Bengal, 741235, India.
- Department of Nanoscience and Nanotechnology, University of Kalyani, Kalyani, West Bengal, 741235, India.
| | - Keka Sarkar
- Department of Microbiology, University of Kalyani, Kalyani, West Bengal, 741235, India.
| |
Collapse
|
3
|
Chan KI, Zhang S, Li G, Xu Y, Cui L, Wang Y, Su H, Tan W, Zhong Z. MYC Oncogene: A Druggable Target for Treating Cancers with Natural Products. Aging Dis 2024; 15:640-697. [PMID: 37450923 PMCID: PMC10917530 DOI: 10.14336/ad.2023.0520] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/24/2023] [Accepted: 05/20/2023] [Indexed: 07/18/2023] Open
Abstract
Various diseases, including cancers, age-associated disorders, and acute liver failure, have been linked to the oncogene, MYC. Animal testing and clinical trials have shown that sustained tumor volume reduction can be achieved when MYC is inactivated, and different combinations of therapeutic agents including MYC inhibitors are currently being developed. In this review, we first provide a summary of the multiple biological functions of the MYC oncoprotein in cancer treatment, highlighting that the equilibrium points of the MYC/MAX, MIZ1/MYC/MAX, and MAD (MNT)/MAX complexes have further potential in cancer treatment that could be used to restrain MYC oncogene expression and its functions in tumorigenesis. We also discuss the multifunctional capacity of MYC in various cellular cancer processes, including its influences on immune response, metabolism, cell cycle, apoptosis, autophagy, pyroptosis, metastasis, angiogenesis, multidrug resistance, and intestinal flora. Moreover, we summarize the MYC therapy patent landscape and emphasize the potential of MYC as a druggable target, using herbal medicine modulators. Finally, we describe pending challenges and future perspectives in biomedical research, involving the development of therapeutic approaches to modulate MYC or its targeted genes. Patients with cancers driven by MYC signaling may benefit from therapies targeting these pathways, which could delay cancerous growth and recover antitumor immune responses.
Collapse
Affiliation(s)
- Ka Iong Chan
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Siyuan Zhang
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Guodong Li
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Yida Xu
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Liao Cui
- Guangdong Provincial Key Laboratory of Research and Development of Natural Drugs, School of Pharmacy, Guangdong Medical University, Zhanjiang 524000, China
| | - Yitao Wang
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Huanxing Su
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| | - Wen Tan
- School of Pharmacy, Lanzhou University, Lanzhou 730000, China
| | - Zhangfeng Zhong
- Macao Centre for Research and Development in Chinese Medicine, State Key Laboratory of Quality Research in Chinese Medicine, Institute of Chinese Medical Sciences, University of Macau, Macao SAR 999078, China
| |
Collapse
|
4
|
Wu S, Edwards H, Wang D, Liu S, Qiao X, Carter J, Wang Y, Taub JW, Wang G, Ge Y. Inhibition of Mcl-1 Synergistically Enhances the Antileukemic Activity of Gilteritinib and MRX-2843 in Preclinical Models of FLT3-Mutated Acute Myeloid Leukemia. Cells 2022; 11:2752. [PMID: 36078163 PMCID: PMC9455003 DOI: 10.3390/cells11172752] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/04/2022] [Revised: 08/25/2022] [Accepted: 09/01/2022] [Indexed: 11/17/2022] Open
Abstract
FMS-like tyrosine kinase 3 (FLT3)-internal tandem duplication (FLT3-ITD) mutations occur in about 25% of all acute myeloid leukemia (AML) patients and confer a poor prognosis. FLT3 inhibitors have been developed to treat patients with FLT3-mutated AML and have shown promise, though the acquisition of resistance occurs, highlighting the need for combination therapies to prolong the response to FLT3 inhibitors. In this study, we investigated the selective Mcl-1 inhibitor AZD5991 in combination with the FLT3 inhibitors gilteritinib and MRX-2843. The combinations synergistically induce apoptosis in AML cell lines and primary patient samples. The FLT3 inhibitors downregulate c-Myc transcripts through the suppression of the MEK/ERK and JAK2/STAT5 pathways, resulting in the decrease in c-Myc protein. This suppression of c-Myc plays an important role in the antileukemic activity of AZD5991. Interestingly, the suppression of c-Myc enhances AZD5991-inudced cytochrome c release and the subsequent induction of apoptosis. AZD5991 enhances the antileukemic activity of the FLT3 inhibitors gilteritinib and MRX-2843 against FLT3-mutated AML in vitro, warranting further development.
Collapse
Affiliation(s)
- Shuangshuang Wu
- Department of Pediatric Hematology, The First Hospital of Jilin University, Changchun 130021, China
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48202, USA
- Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Deying Wang
- The Tumor Center of the First Hospital of Jilin University, Changchun 130021, China
| | - Shuang Liu
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Xinan Qiao
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Jenna Carter
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA
- MD/PhD Program, Wayne State University School of Medicine, Detroit, MI 48201, USA
| | - Yue Wang
- Department of Pediatric Hematology, The First Hospital of Jilin University, Changchun 130021, China
| | - Jeffrey W. Taub
- Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI 48202, USA
- Division of Pediatric Hematology and Oncology, Department of Pediatrics, Children’s Hospital of Michigan, Detroit, MI 48201, USA
- Department of Pediatrics, Central Michigan University College of Medicine, Mt. Pleasant, MI 48859, USA
| | - Guan Wang
- National Engineering Laboratory for AIDS Vaccine, School of Life Sciences, Jilin University, Changchun 130012, China
| | - Yubin Ge
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI 48202, USA
- Molecular Therapeutics Program, Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI 48201, USA
- Cancer Biology Graduate Program, Wayne State University School of Medicine, Detroit, MI 48201, USA
| |
Collapse
|
5
|
Zanotti S, Decaesteker B, Vanhauwaert S, De Wilde B, De Vos WH, Speleman F. Cellular senescence in neuroblastoma. Br J Cancer 2022; 126:1529-1538. [PMID: 35197583 PMCID: PMC9130206 DOI: 10.1038/s41416-022-01755-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/02/2021] [Revised: 01/14/2022] [Accepted: 02/10/2022] [Indexed: 12/14/2022] Open
Abstract
Neuroblastoma is a tumour that arises from the sympathoadrenal lineage occurring predominantly in children younger than five years. About half of the patients are diagnosed with high-risk tumours and undergo intensive multi-modal therapy. The success rate of current treatments for high-risk neuroblastoma is disappointingly low and survivors suffer from multiple therapy-related long-term side effects. Most chemotherapeutics drive cancer cells towards cell death or senescence. Senescence has long been considered to represent a terminal non-proliferative state and therefore an effective barrier against tumorigenesis. This dogma, however, has been challenged by recent observations that infer a much more dynamic and reversible nature for this process, which may have implications for the efficacy of therapy-induced senescence-oriented treatment strategies. Neuroblastoma cells in a dormant, senescent-like state may escape therapy, whilst their senescence-associated secretome may promote inflammation and invasiveness, potentially fostering relapse. Conversely, due to its distinct molecular identity, senescence may also represent an opportunity for the development of novel (combination) therapies. However, the limited knowledge on the molecular dynamics and diversity of senescence signatures demands appropriate models to study this process in detail. This review summarises the molecular knowledge about cellular senescence in neuroblastoma and investigates current and future options towards therapeutic exploration.
Collapse
Affiliation(s)
- Sofia Zanotti
- grid.5284.b0000 0001 0790 3681Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Universiteitsplein 1, Antwerp, 2610 Belgium ,grid.5342.00000 0001 2069 7798Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Ghent, 9000 Belgium ,grid.510942.bCancer Research Institute Ghent (CRIG), Ghent, 9000 Belgium
| | - Bieke Decaesteker
- grid.5342.00000 0001 2069 7798Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Ghent, 9000 Belgium ,grid.510942.bCancer Research Institute Ghent (CRIG), Ghent, 9000 Belgium
| | - Suzanne Vanhauwaert
- grid.5342.00000 0001 2069 7798Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Ghent, 9000 Belgium ,grid.510942.bCancer Research Institute Ghent (CRIG), Ghent, 9000 Belgium
| | - Bram De Wilde
- grid.5342.00000 0001 2069 7798Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Ghent, 9000 Belgium ,grid.5342.00000 0001 2069 7798Department of Internal Medicine and Pediatrics, Ghent University, Corneel Heymanslaan 10, Ghent, 9000 Belgium ,grid.410566.00000 0004 0626 3303Department of Pediatric Hematology Oncology and Stem Cell Transplantation, Ghent University Hospital, Ghent, 9000 Belgium
| | - Winnok H. De Vos
- grid.5284.b0000 0001 0790 3681Laboratory of Cell Biology and Histology, Department of Veterinary Sciences, University of Antwerp, Universiteitsplein 1, Antwerp, 2610 Belgium
| | - Frank Speleman
- Department of Biomolecular Medicine, Ghent University, Corneel Heymanslaan 10, Ghent, 9000, Belgium. .,Cancer Research Institute Ghent (CRIG), Ghent, 9000, Belgium.
| |
Collapse
|
6
|
Donati G, Ravà M, Filipuzzi M, Nicoli P, Cassina L, Verrecchia A, Doni M, Rodighiero S, Parodi F, Boletta A, Vellano CP, Marszalek JR, Draetta GF, Amati B. Targeting mitochondrial respiration and the BCL2 family in high-grade MYC-associated B-cell lymphoma. Mol Oncol 2021; 16:1132-1152. [PMID: 34632715 PMCID: PMC8895457 DOI: 10.1002/1878-0261.13115] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/17/2021] [Revised: 08/27/2021] [Accepted: 10/08/2021] [Indexed: 01/02/2023] Open
Abstract
Multiple molecular features, such as activation of specific oncogenes (e.g., MYC, BCL2) or a variety of gene expression signatures, have been associated with disease course in diffuse large B‐cell lymphoma (DLBCL), although their relationships and implications for targeted therapy remain to be fully unraveled. We report that MYC activity is closely correlated with—and most likely a driver of—gene signatures related to oxidative phosphorylation (OxPhos) in DLBCL, pointing to OxPhos enzymes, in particular mitochondrial electron transport chain (ETC) complexes, as possible therapeutic targets in high‐grade MYC‐associated lymphomas. In our experiments, indeed, MYC sensitized B cells to the ETC complex I inhibitor IACS‐010759. Mechanistically, IACS‐010759 triggered the integrated stress response (ISR) pathway, driven by the transcription factors ATF4 and CHOP, which engaged the intrinsic apoptosis pathway and lowered the apoptotic threshold in MYC‐overexpressing cells. In line with these findings, the BCL2‐inhibitory compound venetoclax synergized with IACS‐010759 against double‐hit lymphoma (DHL), a high‐grade malignancy with concurrent activation of MYC and BCL2. In BCL2‐negative lymphoma cells, instead, killing by IACS‐010759 was potentiated by the Mcl‐1 inhibitor S63845. Thus, combining an OxPhos inhibitor with select BH3‐mimetic drugs provides a novel therapeutic principle against aggressive, MYC‐associated DLBCL variants.
Collapse
Affiliation(s)
- Giulio Donati
- European Institute of Oncology (IEO)-IRCCS, Milan, Italy
| | - Micol Ravà
- European Institute of Oncology (IEO)-IRCCS, Milan, Italy
| | | | - Paola Nicoli
- European Institute of Oncology (IEO)-IRCCS, Milan, Italy
| | - Laura Cassina
- IRCCS San Raffaele Scientific Institute, Milan, Italy
| | | | - Mirko Doni
- European Institute of Oncology (IEO)-IRCCS, Milan, Italy
| | | | | | | | - Christopher P Vellano
- Translational Research to Advance Therapeutics and Innovation in Oncology (TRACTION), Houston, TX, USA
| | - Joseph R Marszalek
- Translational Research to Advance Therapeutics and Innovation in Oncology (TRACTION), Houston, TX, USA
| | - Giulio F Draetta
- Department of Genomic Medicine, University of Texas MD Anderson Cancer Center, Houston, TX, USA
| | - Bruno Amati
- European Institute of Oncology (IEO)-IRCCS, Milan, Italy
| |
Collapse
|
7
|
Ahmadi SE, Rahimi S, Zarandi B, Chegeni R, Safa M. MYC: a multipurpose oncogene with prognostic and therapeutic implications in blood malignancies. J Hematol Oncol 2021; 14:121. [PMID: 34372899 PMCID: PMC8351444 DOI: 10.1186/s13045-021-01111-4] [Citation(s) in RCA: 102] [Impact Index Per Article: 25.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2021] [Accepted: 06/12/2021] [Indexed: 12/17/2022] Open
Abstract
MYC oncogene is a transcription factor with a wide array of functions affecting cellular activities such as cell cycle, apoptosis, DNA damage response, and hematopoiesis. Due to the multi-functionality of MYC, its expression is regulated at multiple levels. Deregulation of this oncogene can give rise to a variety of cancers. In this review, MYC regulation and the mechanisms by which MYC adjusts cellular functions and its implication in hematologic malignancies are summarized. Further, we also discuss potential inhibitors of MYC that could be beneficial for treating hematologic malignancies.
Collapse
Affiliation(s)
- Seyed Esmaeil Ahmadi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Samira Rahimi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Bahman Zarandi
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran
| | - Rouzbeh Chegeni
- Medical Laboratory Sciences Program, College of Health and Human Sciences, Northern Illinois University, DeKalb, IL, USA.
| | - Majid Safa
- Department of Hematology and Blood Banking, Faculty of Allied Medicine, Iran University of Medical Sciences, Tehran, Iran.
- Cellular and Molecular Research Center, Iran University of Medical Sciences, Tehran, Iran.
| |
Collapse
|
8
|
Wartalski K, Gorczyca G, Wiater J, Tabarowski Z, Duda M. Porcine ovarian cortex-derived putative stem cells can differentiate into endothelial cells in vitro. Histochem Cell Biol 2021; 156:349-362. [PMID: 34269874 PMCID: PMC8550686 DOI: 10.1007/s00418-021-02016-6] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 07/05/2021] [Indexed: 12/19/2022]
Abstract
Endothelial cells (ECs), the primary component of the vasculature, play a crucial role in neovascularization. However, the number of endogenous ECs is inadequate for both experimental purposes and clinical applications. Porcine ovarian putative stem cells (poPSCs), although not pluripotent, are characterized by great plasticity. Therefore, this study aimed to investigate whether poPSCs have the potential to differentiate into cells of endothelial lineage. poPSCs were immunomagnetically isolated from postnatal pig ovaries based on the presence of SSEA-4 protein. Expression of mesenchymal stem cells (MSCs) markers after pre-culture, both at the level of mRNA: ITGB1, THY, and ENG and corresponding protein: CD29, CD90, and CD105 were significantly higher compared to the control ovarian cortex cells. To differentiate poPSCs into ECs, inducing medium containing vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), insulin-like growth factor (IGF), epidermal growth factor (EGF), ascorbic acid, and heparin was applied. After 14 days, poPSC differentiation into ECs was confirmed by immunofluorescence staining for vascular endothelial cadherin (VECad) and vascular endothelial growth factor receptor-2 (VEGFR-2). Semi-quantitative WB analysis of these proteins confirmed their high abundance. Additionally, qRT-PCR showed that mRNA expression of corresponding marker genes: CDH5, KDR was significantly higher compared with undifferentiated poPSCs. Finally, EC functional status was confirmed by the migration test that revealed that they were capable of positive chemotaxis, while tube formation assay demonstrated their ability to develop capillary networks. In conclusion, our results provided evidence that poPSCs may constitute the MSC population in the ovary and confirmed that they might be a potential source of ECs for tissue engineering.
Collapse
Affiliation(s)
- Kamil Wartalski
- Faculty of Medicine, Department of Histology, Jagiellonian University Medical College, Kopernika 7 Street, 31-034, Krakow, Poland
| | - Gabriela Gorczyca
- Faculty of Biology, Institute of Zoology and Biomedical Research, Department of Endocrinology, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387, Krakow, Poland
| | - Jerzy Wiater
- Faculty of Medicine, Department of Histology, Jagiellonian University Medical College, Kopernika 7 Street, 31-034, Krakow, Poland
| | - Zbigniew Tabarowski
- Faculty of Biology, Institute of Zoology and Biomedical Research, Department of Experimental Hematology, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387, Krakow, Poland
| | - Małgorzata Duda
- Faculty of Biology, Institute of Zoology and Biomedical Research, Department of Endocrinology, Jagiellonian University in Krakow, Gronostajowa 9 Street, 30-387, Krakow, Poland.
| |
Collapse
|
9
|
Sabit H, Tombuloglu H, Cevik E, Abdel-Ghany S, El-Zawahri E, El-Sawy A, Isik S, Al-Suhaimi E. Knockdown of c-MYC Controls the Proliferation of Oral Squamous Cell Carcinoma Cells in vitro via Dynamic Regulation of Key Apoptotic Marker Genes. INTERNATIONAL JOURNAL OF MOLECULAR AND CELLULAR MEDICINE 2021; 10:45-55. [PMID: 34268253 PMCID: PMC8256829 DOI: 10.22088/ijmcm.bums.10.1.45] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 09/24/2020] [Accepted: 05/09/2021] [Indexed: 01/09/2023]
Abstract
Oral squamous cell carcinoma (OSCC) is the most common malignant epithelial cancer occurring in the oral cavity, where it accounts for nearly 90% of all oral cavity neoplasms. The c-MYC transcription factor plays an important role in the control of programmed cell death, normal-to-malignant cellular transformation, and progression of the cell cycle. However, the role of c-MYC in controlling the proliferation of OSCC cells is not well known. In this study, c-MYC gene was silenced in OSCC cells (ORL-136T), and molecular and cellular responses were screened. To identify the pathway through which cell death occurred, cytotoxicity, colony formation, western blotting, caspase-3, and RT-qPCR analyzes were performed. Results indicated that knockdown of c-MYC has resulted in a significant decrease in the cell viability and c-MYC protein synthesis. Furthermore, caspase-3 was shown to be upregulated leading to apoptosis via the intrinsic pathway. In response to c-MYC knockdown, eight cell proliferation-associated genes showed variable expression profiles: c-MYC (-21.2), p21 (-2.5), CCNA1(1.8), BCL2 (-1.4), p53(-3.7), BAX(1.1), and CYCS (19.3). p27 expression was dramatically decreased in c-MYC-silenced cells in comparison with control, and this might indicate that the relative absence of c-MYC triggered intrinsic apoptosis in OSCC cells via p27 and CYCS.
Collapse
Affiliation(s)
- Hussein Sabit
- Department of Genetics, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.
| | - Huseyin Tombuloglu
- Department of Genetics, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.
| | - Emre Cevik
- Department of Genetics, Institute for Research and Medical Consultations, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.
| | - Shaimaa Abdel-Ghany
- College of Biotechnology, Misr University for Science and Technology, Giza, Egypt.
| | - Engy El-Zawahri
- College of Biotechnology, Misr University for Science and Technology, Giza, Egypt.
| | - Amr El-Sawy
- College of Biotechnology, Misr University for Science and Technology, Giza, Egypt.
| | - Sevim Isik
- Department of Molecular Biology and Genetics, Faculty of Engineering and Natural Sciences, Uskudar University, Istanbul, Turkey.
- SANKARA Brain & Biotechnology Research Center, Istanbul Biotechnology Inc, Technocity, Avcilar, Istanbul, Turkey.
| | - Ebtesam Al-Suhaimi
- Department of Biology, College of Science, Imam Abdulrahman Bin Faisal University, Dammam, Saudi Arabia.
| |
Collapse
|
10
|
Yan J, Xie Y, Si J, Gan L, Li H, Sun C, Di C, Zhang J, Huang G, Zhang X, Zhang H. Crosstalk of the Caspase Family and Mammalian Target of Rapamycin Signaling. Int J Mol Sci 2021; 22:E817. [PMID: 33467535 PMCID: PMC7830632 DOI: 10.3390/ijms22020817] [Citation(s) in RCA: 11] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2020] [Revised: 01/11/2021] [Accepted: 01/12/2021] [Indexed: 12/20/2022] Open
Abstract
Cell can integrate the caspase family and mammalian target of rapamycin (mTOR) signaling in response to cellular stress triggered by environment. It is necessary here to elucidate the direct response and interaction mechanism between the two signaling pathways in regulating cell survival and determining cell fate under cellular stress. Members of the caspase family are crucial regulators of inflammation, endoplasmic reticulum stress response and apoptosis. mTOR signaling is known to mediate cell growth, nutrition and metabolism. For instance, over-nutrition can cause the hyperactivation of mTOR signaling, which is associated with diabetes. Nutrition deprivation can inhibit mTOR signaling via SH3 domain-binding protein 4. It is striking that Ras GTPase-activating protein 1 is found to mediate cell survival in a caspase-dependent manner against increasing cellular stress, which describes a new model of apoptosis. The components of mTOR signaling-raptor can be cleaved by caspases to control cell growth. In addition, mTOR is identified to coordinate the defense process of the immune system by suppressing the vitality of caspase-1 or regulating other interferon regulatory factors. The present review discusses the roles of the caspase family or mTOR pathway against cellular stress and generalizes their interplay mechanism in cell fate determination.
Collapse
Affiliation(s)
- Junfang Yan
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Yi Xie
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Jing Si
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Lu Gan
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Hongyan Li
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Chao Sun
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Cuixia Di
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| | - Jinhua Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Guomin Huang
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Xuetian Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
- College of Life Sciences, University of Chinese Academy of Sciences, Beijing 101408, China
| | - Hong Zhang
- Institute of Modern Physics, Chinese Academy of Sciences, 509 Nanchang Road, Lanzhou 730000, China; (J.Y.); (J.S.); (L.G.); (H.L.); (C.S.); (C.D.); (J.Z.); (G.H.); (X.Z.)
- Advanced Energy Science and Technology Guangdong Laboratory, Huizhou 516029, China
- Key Laboratory of Heavy Ion Radiation Biology and Medicine of Chinese Academy of Sciences, Lanzhou 730000, China
- Key Laboratory of Heavy Ion Radiation Medicine of Gansu Province, Lanzhou 730000, China
| |
Collapse
|
11
|
Lu D, Wilson C, Littlewood TD. Methods for Determining Myc-Induced Apoptosis. Methods Mol Biol 2021; 2318:209-229. [PMID: 34019292 DOI: 10.1007/978-1-0716-1476-1_10] [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] [Indexed: 02/11/2024]
Abstract
Although many oncoproteins promote cell growth and proliferation, some also possess the potential to induce cell cycle arrest or cell death by apoptosis. Elevated and deregulated expression of the Myc protein promotes apoptosis in both cultured cells and in some tissues in vivo. Here we describe techniques to detect Myc-induced apoptosis in vitro using flow cytometry, microscopy, and immunoblotting, and in vivo using immunohistochemical staining, immunoblotting, and analysis of RNA expression.
Collapse
Affiliation(s)
- Dan Lu
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | - Catherine Wilson
- Department of Pharmacology, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
12
|
Choi SH, Koh DI, Ahn H, Kim JY, Kim Y, Hur MW. Cell fate decisions by c-Myc depend on ZBTB5 and p53. Biochem Biophys Res Commun 2020; 533:1247-1254. [PMID: 33051058 DOI: 10.1016/j.bbrc.2020.09.137] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2020] [Revised: 09/29/2020] [Accepted: 09/29/2020] [Indexed: 11/17/2022]
Abstract
The oncoprotein, c-Myc, not only promotes cell proliferation, but can also induce or sensitize cells to apoptosis. However, how c-Myc decides cell fate and which c-Myc downstream target genes are involved remain unknown. Previously, we showed that ZBTB5 (zinc finger and BTB domain-containing 5) is a proto-oncogene that stimulates cell proliferation. ZBTB5 represses p21/CDKN1A by competing with p53 and recruiting corepressor histone deacetylase complexes. Herein, we found that c-Myc directly activates the transcription of ZBTB5. In the absence of p53, ZBTB5 is acetylated at K597 by interacting with p300, and activates transcription of NOXA, which induces apoptosis. In contrast, in the presence of p53, ZBTB5 interacts with p53 and acetylation at ZBTB5 K597 is blocked. ZBTB5 without K597 acetylation interacts with mSin3A/HDAC1 to repress p21/CDKN1A transcription and promote cell proliferation. Cell fate decisions by c-Myc depend on ZBTB5, p53 and p300, and acetylation of ZBTB5 K597.
Collapse
Affiliation(s)
- Seo-Hyun Choi
- Brain Korea 21 Plus Project for Medical Science, Severance Biomedical Research Institute, Department of Biochemistry and Molecular Biology, Yonsei University School of Medicine, 50-1 Yonsei-Ro, SeoDaeMoon-Ku, Seoul, 03722, South Korea
| | - Dong-In Koh
- Brain Korea 21 Plus Project for Medical Science, Severance Biomedical Research Institute, Department of Biochemistry and Molecular Biology, Yonsei University School of Medicine, 50-1 Yonsei-Ro, SeoDaeMoon-Ku, Seoul, 03722, South Korea
| | - Haemin Ahn
- Brain Korea 21 Plus Project for Medical Science, Severance Biomedical Research Institute, Department of Biochemistry and Molecular Biology, Yonsei University School of Medicine, 50-1 Yonsei-Ro, SeoDaeMoon-Ku, Seoul, 03722, South Korea
| | - Jin Young Kim
- Mass Spectrometry Research Center, Korea Basic Science Institute, 804-1 Yangcheong-ri, Ochang-eup, Cheongwon-gun, Chungbuk, 34133, South Korea
| | - Youngsoo Kim
- Department of Biomedical Sciences and Biomedical Engineering, Seoul National University College of Medicine, Seoul, 03080, South Korea
| | - Man-Wook Hur
- Brain Korea 21 Plus Project for Medical Science, Severance Biomedical Research Institute, Department of Biochemistry and Molecular Biology, Yonsei University School of Medicine, 50-1 Yonsei-Ro, SeoDaeMoon-Ku, Seoul, 03722, South Korea.
| |
Collapse
|
13
|
Chowdhury MMR, Park J, Afrin F, Ko YG, Kim CL, Lee SS, Kim SW. Transcriptome profiling of in vitro-matured oocytes from a korean native cow (hanwoo) after cysteamine supplementation. Anim Biotechnol 2020; 32:401-412. [PMID: 31900040 DOI: 10.1080/10495398.2019.1706545] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
This study elucidated the molecular markers that decrease oocyte quality during in vitro culture, restricting optimal developmental potential. Here, we evaluated the transcriptomic differences between cysteamine-treated and non-treated bovine cumulus oocyte complexes (COCs) after 22 h of co-culture in the maturation media using RNA sequencing. In total, 39,014 transcripts were sequenced between cysteamine-treated and non-treated mature COCs. We evaluated the relative expression of 21,472 genes, with 59 genes showing differential expression between the two COC groups. The cysteamine-treated group had 36 up-regulated gene transcripts and 23 down-regulated gene transcripts. Moreover, gene ontology (GO) enrichment analysis revealed that multiple biological processes were significantly enriched after cysteamine supplementation. Differentially expressed genes appeared to maintain normal oocyte physiology, regulation of apoptosis, differentiation, ossification or bone formation, cardiac and muscle physiology, hormonal secretion, and membrane construction for further embryonic development. In conclusion, cysteamine affects the mRNA level of COCs during oocyte maturation by upregulating potential molecular markers and downregulating genes that affect further embryonic development.
Collapse
Affiliation(s)
- M M R Chowdhury
- Animal Genetic Resources Research Center, National Institute of Animal Science (NIAS), Rural Development Administration (RDA), Hamyang, Republic of Korea.,Department of Physiology and Pharmacology, Faculty of Animal Science and Veterinary Medicine, Patuakhali Science and Technology University, Dhaka, Bangladesh
| | - Joonghoon Park
- Department of International Agricultural Technology, Graduate School of International Agricultural Technology, Seoul National University, Pyeongchang, Republic of Korea.,Institute of Green Bio Science and Technology, Seoul National University, Pyeongchang, Republic of Korea
| | - Fahmida Afrin
- Animal Genetic Resources Research Center, National Institute of Animal Science (NIAS), Rural Development Administration (RDA), Hamyang, Republic of Korea
| | - Yeoung-Gyu Ko
- Animal Genetic Resources Research Center, National Institute of Animal Science (NIAS), Rural Development Administration (RDA), Hamyang, Republic of Korea
| | - Chan-Lan Kim
- Animal Genetic Resources Research Center, National Institute of Animal Science (NIAS), Rural Development Administration (RDA), Hamyang, Republic of Korea
| | - Sung Soo Lee
- Animal Genetic Resources Research Center, National Institute of Animal Science (NIAS), Rural Development Administration (RDA), Hamyang, Republic of Korea
| | - Sung Woo Kim
- Animal Genetic Resources Research Center, National Institute of Animal Science (NIAS), Rural Development Administration (RDA), Hamyang, Republic of Korea
| |
Collapse
|
14
|
Khaiboullina S, Uppal T, Kletenkov K, St Jeor SC, Garanina E, Rizvanov A, Verma SC. Transcriptome Profiling Reveals Pro-Inflammatory Cytokines and Matrix Metalloproteinase Activation in Zika Virus Infected Human Umbilical Vein Endothelial Cells. Front Pharmacol 2019; 10:642. [PMID: 31249527 PMCID: PMC6582368 DOI: 10.3389/fphar.2019.00642] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/07/2018] [Accepted: 05/17/2019] [Indexed: 12/19/2022] Open
Abstract
The deformities in the newborns infected with Zika virus (ZIKV) present a new potential public health threat to the worldwide community. Although ZIKV infection is mainly asymptomatic in healthy adults, infection during pregnancy can cause microcephaly and other severe brain defects and potentially death of the fetus. The detailed mechanism of ZIKV-associated damage is still largely unknown; however, it is apparent that the virus crosses the placental barrier to reach the fetus. Endothelial cells are the key structural component of the placental barrier. Endothelium integrity as semi-permeable barrier is essential to control the molecules and leukocytes trafficking across the placenta. Damaged endothelium or disruption of adherens junctions could compromise endothelial barrier integrity causing leakage and inflammation. Endothelial cells are often targeted by viruses, including the members of the Flaviviridae family such as dengue virus (DENV) and West Nile virus (WNV); however, little is known about the effects of ZIKV infection of endothelial cell functions. Our transcriptomic data have demonstrated that the large number of cytokines is affected in ZIKV-infected endothelial cells, where significant changes in 13 and 11 cytokines were identified in cells infected with PRVABC59 and IBH30656 ZIKV strains, respectively. Importantly, these cytokines include chemokines attracting mononuclear leukocytes (monocytes and lymphocytes) as well as neutrophils. Additionally, changes in matrix metalloproteinase (MMPs) were detected in ZIKV-infected cells. Furthermore, we for the first time showed that ZIKV infection of human umbilical vein endothelial cells (HUVECs) increases endothelial permeability. We reason that increased endothelial permeability was due to apoptosis of endothelial cells caused by caspase-8 activation in ZIKV-infected cells.
Collapse
Affiliation(s)
- Svetlana Khaiboullina
- Department of Microbiology and Immunology, University of Nevada, Reno, Reno, NV, United States
| | - Timsy Uppal
- Department of Microbiology and Immunology, University of Nevada, Reno, Reno, NV, United States
| | - Konstatin Kletenkov
- Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Kazan Federal University, Kazan, Russia
| | - Stephen Charles St Jeor
- Department of Microbiology and Immunology, University of Nevada, Reno, Reno, NV, United States.,Genequest LLC, Reno, NV, United States
| | - Ekaterina Garanina
- Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Kazan Federal University, Kazan, Russia
| | - Albert Rizvanov
- Department of Exploratory Research, Scientific and Educational Center of Pharmaceutics, Kazan Federal University, Kazan, Russia
| | - Subhash C Verma
- Department of Microbiology and Immunology, University of Nevada, Reno, Reno, NV, United States
| |
Collapse
|
15
|
Mayers S, Moço PD, Maqbool T, Silva PN, Kilkenny DM, Audet J. Establishment of an erythroid progenitor cell line capable of enucleation achieved with an inducible c-Myc vector. BMC Biotechnol 2019; 19:21. [PMID: 30987611 PMCID: PMC6466758 DOI: 10.1186/s12896-019-0515-9] [Citation(s) in RCA: 7] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/17/2018] [Accepted: 04/05/2019] [Indexed: 01/13/2023] Open
Abstract
BACKGROUND A robust scalable method for producing enucleated red blood cells (RBCs) is not only a process to produce packed RBC units for transfusion but a potential platform to produce modified RBCs with applications in advanced cellular therapy. Current strategies for producing RBCs have shortcomings in the limited self-renewal capacity of progenitor cells, or difficulties in effectively enucleating erythroid cell lines. We explored a new method to produce RBCs by inducibly expressing c-Myc in primary erythroid progenitor cells and evaluated the proliferative and maturation potential of these modified cells. RESULTS Primary erythroid progenitor cells were genetically modified with an inducible gene transfer vector expressing a single transcription factor, c-Myc, and all the gene elements required to achieve dox-inducible expression. Genetically modified cells had enhanced proliferative potential compared to control cells, resulting in exponential growth for at least 6 weeks. Inducibly proliferating erythroid (IPE) cells were isolated with surface receptors similar to colony forming unit-erythroid (CFU-Es), and after removal of ectopic c-Myc expression cells hemoglobinized, decreased in cell size to that of native RBCs, and enucleated achieving cultures with 17% enucleated cells. Experiments with IPE cells at various levels of ectopic c-Myc expression provided insight into differentiation dynamics of the modified cells, and an optimized two-stage differentiation strategy was shown to promote greater expansion and maturation. CONCLUSIONS Genetic engineering of adult erythroid progenitor cells with an inducible c-Myc vector established an erythroid progenitor cell line that could produce RBCs, demonstrating the potential of this approach to produce large quantities of RBCs and modified RBC products.
Collapse
Affiliation(s)
- Steven Mayers
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Canada.,Institute of Biomaterials and Biomedical Engineering (IBBME), University of Toronto, Toronto, Canada
| | - Pablo Diego Moço
- Institute of Biomaterials and Biomedical Engineering (IBBME), University of Toronto, Toronto, Canada
| | - Talha Maqbool
- Institute of Biomaterials and Biomedical Engineering (IBBME), University of Toronto, Toronto, Canada
| | - Pamuditha N Silva
- Institute of Biomaterials and Biomedical Engineering (IBBME), University of Toronto, Toronto, Canada
| | - Dawn M Kilkenny
- Institute of Biomaterials and Biomedical Engineering (IBBME), University of Toronto, Toronto, Canada
| | - Julie Audet
- Department of Chemical Engineering and Applied Chemistry, University of Toronto, Toronto, Canada. .,Institute of Biomaterials and Biomedical Engineering (IBBME), University of Toronto, Toronto, Canada.
| |
Collapse
|
16
|
Romeo M, Hutchison T, Malu A, White A, Kim J, Gardner R, Smith K, Nelson K, Bergeson R, McKee R, Harrod C, Ratner L, Lüscher B, Martinez E, Harrod R. The human T-cell leukemia virus type-1 p30 II protein activates p53 and induces the TIGAR and suppresses oncogene-induced oxidative stress during viral carcinogenesis. Virology 2018; 518:103-115. [PMID: 29462755 DOI: 10.1016/j.virol.2018.02.010] [Citation(s) in RCA: 14] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2017] [Revised: 01/23/2018] [Accepted: 02/09/2018] [Indexed: 01/08/2023]
Abstract
In normal cells, aberrant oncogene expression leads to the accumulation of cytotoxic metabolites, including reactive oxygen species (ROS), which can cause oxidative DNA-damage and apoptosis as an intrinsic barrier against neoplastic disease. The c-Myc oncoprotein is overexpressed in many lymphoid cancers due to c-myc gene amplification and/or 8q24 chromosomal translocations. Intriguingly, p53 is a downstream target of c-Myc and hematological malignancies, such as adult T-cell leukemia/lymphoma (ATL), frequently contain wildtype p53 and c-Myc overexpression. We therefore hypothesized that p53-regulated pro-survival signals may thwart the cell's metabolic anticancer defenses to support oncogene-activation in lymphoid cancers. Here we show that the Tp53-induced glycolysis and apoptosis regulator (TIGAR) promotes c-myc oncogene-activation by the human T-cell leukemia virus type-1 (HTLV-1) latency-maintenance factor p30II, associated with c-Myc deregulation in ATL clinical isolates. TIGAR prevents the intracellular accumulation of c-Myc-induced ROS and inhibits oncogene-induced cellular senescence in ATL, acute lymphoblastic leukemia, and multiple myeloma cells with elevated c-Myc expression. Our results allude to a pivotal role for p53-regulated antioxidant signals as mediators of c-Myc oncogenic functions in viral and non-viral lymphoid tumors.
Collapse
Affiliation(s)
- Megan Romeo
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX 75275-0376, Unites States
| | - Tetiana Hutchison
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX 75275-0376, Unites States
| | - Aditi Malu
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX 75275-0376, Unites States
| | - Averi White
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX 75275-0376, Unites States
| | - Janice Kim
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX 75275-0376, Unites States
| | - Rachel Gardner
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX 75275-0376, Unites States
| | - Katie Smith
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX 75275-0376, Unites States
| | - Katherine Nelson
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX 75275-0376, Unites States
| | - Rachel Bergeson
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX 75275-0376, Unites States
| | - Ryan McKee
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX 75275-0376, Unites States
| | - Carolyn Harrod
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX 75275-0376, Unites States
| | - Lee Ratner
- Departments of Medicine and Molecular Microbiology, Washington University School of Medicine, St. Louis, MO 63110, United States
| | - Bernhard Lüscher
- Institute of Biochemistry, Klinikum, RWTH Aachen University, Pauwelsstrasse 30, 52057 Aachen, Germany
| | - Ernest Martinez
- Department of Biochemistry, University of California, Riverside, CA 92521, United States
| | - Robert Harrod
- Laboratory of Molecular Virology, Department of Biological Sciences, and The Dedman College Center for Drug Discovery, Design & Delivery, Southern Methodist University, Dallas, TX 75275-0376, Unites States.
| |
Collapse
|
17
|
BCL-X L directly modulates RAS signalling to favour cancer cell stemness. Nat Commun 2017; 8:1123. [PMID: 29066722 PMCID: PMC5654832 DOI: 10.1038/s41467-017-01079-1] [Citation(s) in RCA: 41] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/02/2017] [Accepted: 08/16/2017] [Indexed: 11/08/2022] Open
Abstract
In tumours, accumulation of chemoresistant cells that express high levels of anti-apoptotic proteins such as BCL-XL is thought to result from the counter selection of sensitive, low expresser clones during progression and/or initial treatment. We herein show that BCL-XL expression is selectively advantageous to cancer cell populations even in the absence of pro-apoptotic pressure. In transformed human mammary epithelial cells BCL-XL favours full activation of signalling downstream of constitutively active RAS with which it interacts in a BH4-dependent manner. Comparative proteomic analysis and functional assays indicate that this is critical for RAS-induced expression of stemness regulators and maintenance of a cancer initiating cell (CIC) phenotype. Resistant cancer cells thus arise from a positive selection driven by BCL-XL modulation of RAS-induced self-renewal, and during which apoptotic resistance is not necessarily the directly selected trait. BCL-XL provides a survival advantage to cancer cells even in the absence of apoptotic pressures. In this study, the authors show that BCL-XL interacts with RAS in a BH4-dependent manner and regulates RAS-mediated activation of pathways involved in the stemness feature of breast cancer cells.
Collapse
|
18
|
Sipos F, Firneisz G, Műzes G. Therapeutic aspects of c-MYC signaling in inflammatory and cancerous colonic diseases. World J Gastroenterol 2016; 22:7938-7950. [PMID: 27672289 PMCID: PMC5028808 DOI: 10.3748/wjg.v22.i35.7938] [Citation(s) in RCA: 68] [Impact Index Per Article: 7.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/07/2016] [Revised: 08/04/2016] [Accepted: 08/23/2016] [Indexed: 02/06/2023] Open
Abstract
Colonic inflammation is required to heal infections, wounds, and maintain tissue homeostasis. As the seventh hallmark of cancer, however, it may affect all phases of tumor development, including tumor initiation, promotion, invasion and metastatic dissemination, and also evasion immune surveillance. Inflammation acts as a cellular stressor and may trigger DNA damage or genetic instability, and, further, chronic inflammation can provoke genetic mutations and epigenetic mechanisms that promote malignant cell transformation. Both sporadical and colitis-associated colorectal carcinogenesis are multi-step, complex processes arising from the uncontrolled proliferation and spreading of malignantly transformed cell clones with the obvious ability to evade the host's protective immunity. In cells upon DNA damage several proto-oncogenes, including c-MYC are activated in parelell with the inactivation of tumor suppressor genes. The target genes of the c-MYC protein participate in different cellular functions, including cell cycle, survival, protein synthesis, cell adhesion, and micro-RNA expression. The transcriptional program regulated by c-MYC is context dependent, therefore the final cellular response to elevated c-MYC levels may range from increased proliferation to augmented apoptosis. Considering physiological intestinal homeostasis, c-MYC displays a fundamental role in the regulation of cell proliferation and crypt cell number. However, c-MYC gene is frequently deregulated in inflammation, and overexpressed in both sporadic and colitis-associated colon adenocarcinomas. Recent results demonstrated that endogenous c-MYC is essential for efficient induction of p53-dependent apoptosis following DNA damage, but c-MYC function is also involved in and regulated by autophagy-related mechanisms, while its expression is affected by DNA-methylation, or histone acetylation. Molecules directly targeting c-MYC, or agents acting on other genes involved in the c-MYC pathway could be selected for combined regiments. However, due to its context-dependent cellular function, it is clinically essential to consider which cytotoxic drugs are used in combination with c-MYC targeted agents in various tissues. Increasing our knowledge about MYC-dependent pathways might provide direction to novel anti-inflammatory and colorectal cancer therapies.
Collapse
|
19
|
Wang X, Martínez MA, Cheng G, Liu Z, Huang L, Dai M, Chen D, Martínez-Larrañaga MR, Anadón A, Yuan Z. The critical role of oxidative stress in the toxicity and metabolism of quinoxaline 1,4-di-N-oxides in vitro and in vivo. Drug Metab Rev 2016; 48:159-82. [PMID: 27285897 DOI: 10.1080/03602532.2016.1189560] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Quinoxaline 1,4-dioxide derivatives (QdNOs) have been widely used as growth promoters and antibacterial agents. Carbadox (CBX), olaquindox (OLA), quinocetone (QCT), cyadox (CYA) and mequindox (MEQ) are the classical members of QdNOs. Some members of QdNOs are known to cause a variety of toxic effects. To date, however, almost no review has addressed the toxicity and metabolism of QdNOs in relation to oxidative stress. This review focused on the research progress associated with oxidative stress as a plausible mechanism for QdNO-induced toxicity and metabolism. The present review documented that the studies were performed over the past 10 years to interpret the generation of reactive oxygen species (ROS) and oxidative stress as the results of QdNO treatment and have correlated them with various types of QdNO toxicity, suggesting that oxidative stress plays critical roles in their toxicities. The major metabolic pathways of QdNOs are N→O group reduction and hydroxylation. Xanthine oxidoreductase (XOR), aldehyde oxidase (SsAOX1), carbonyl reductase (CBR1) and cytochrome P450 (CYP) enzymes were involved in the QdNOs metabolism. Further understanding the role of oxidative stress in QdNOs-induced toxicity will throw new light onto the use of antioxidants and scavengers of ROS as well as onto the blind spots of metabolism and the metabolizing enzymes of QdNOs. The present review might contribute to revealing the QdNOs toxicity, protecting against oxidative damage and helping to improve the rational use of concurrent drugs, while developing novel QdNO compounds with more efficient potentials and less toxic effects.
Collapse
Affiliation(s)
- Xu Wang
- a National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues , Wuhan , Hubei , China ;,b Department of Toxicology and Pharmacology, Faculty of Veterinary Medicine , Universidad Complutense de Madrid , Madrid , Spain
| | - María-Aránzazu Martínez
- b Department of Toxicology and Pharmacology, Faculty of Veterinary Medicine , Universidad Complutense de Madrid , Madrid , Spain
| | - Guyue Cheng
- c MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products , Huazhong Agricultural University , Wuhan , Hubei , China
| | - Zhaoying Liu
- d Hunan Engineering Research Center of Veterinary Drugs, College of Veterinary Medicine , Hunan Agricultural University , Changsha , Hunan , China
| | - Lingli Huang
- c MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products , Huazhong Agricultural University , Wuhan , Hubei , China
| | - Menghong Dai
- c MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products , Huazhong Agricultural University , Wuhan , Hubei , China
| | - Dongmei Chen
- c MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products , Huazhong Agricultural University , Wuhan , Hubei , China
| | - María-Rosa Martínez-Larrañaga
- b Department of Toxicology and Pharmacology, Faculty of Veterinary Medicine , Universidad Complutense de Madrid , Madrid , Spain
| | - Arturo Anadón
- b Department of Toxicology and Pharmacology, Faculty of Veterinary Medicine , Universidad Complutense de Madrid , Madrid , Spain
| | - Zonghui Yuan
- a National Reference Laboratory of Veterinary Drug Residues (HZAU) and MAO Key Laboratory for Detection of Veterinary Drug Residues , Wuhan , Hubei , China ;,c MOA Laboratory for Risk Assessment of Quality and Safety of Livestock and Poultry Products , Huazhong Agricultural University , Wuhan , Hubei , China ;,e Hubei Collaborative Innovation Center for Animal Nutrition and Feed Safety , Wuhan , Hubei , China
| |
Collapse
|
20
|
Cao W, Wei W, Zhan Z, Xie Y, Xiao Q. MiR-1284 modulates multidrug resistance of gastric cancer cells by targeting EIF4A1. Oncol Rep 2016; 35:2583-91. [PMID: 26936591 DOI: 10.3892/or.2016.4643] [Citation(s) in RCA: 35] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2015] [Accepted: 12/18/2015] [Indexed: 12/11/2022] Open
Abstract
Routine chemotherapy as an important treatment mode often can not be effective because of multidrug resistance (MDR). MicroRNA (miRNA) modulates the expression of a great number of genes, including MDR. In this study, the expression of miR-1284 was reduced in gastric cancer (GC) tissue specimens with metastasis and in vincristine-resistant (VCR) GC SGC7901 cells (SGC-7901/VCR) compared to that in the controls. Recombinant lentiviral vectors with miR-1284 led to the overexpression of miR-1284 mRNA and reversed the chemoresistance of SGC7901/VCR cells, promoted cell cycle arrested at the G0/G1 phase, accelerated drug-induced apoptosis, and decreased migration and invasiveness of SGC-7901/VCR. In addition, the overexpression of miR-1284 sensitized tumors to chemotherapy in vivo. Our data provide combined evidence that miR-1284 can heighten the expression of MYC and reduce the expression of JUN, MMP12, and EIF4A1 that was the direct target. In conclusion, miR-1284 can function as a new regulator to reduce GC MDR cells by targeting EIF4A1.
Collapse
Affiliation(s)
- Wenlong Cao
- Department of Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Weiyuan Wei
- Department of Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Zexu Zhan
- Department of Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Yubo Xie
- Department of Anaesthesiology, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| | - Qiang Xiao
- Department of Surgery, The First Affiliated Hospital of Guangxi Medical University, Nanning, Guangxi 530021, P.R. China
| |
Collapse
|
21
|
Guo H, Chen L, Cui H, Peng X, Fang J, Zuo Z, Deng J, Wang X, Wu B. Research Advances on Pathways of Nickel-Induced Apoptosis. Int J Mol Sci 2015; 17:E10. [PMID: 26703593 PMCID: PMC4730257 DOI: 10.3390/ijms17010010] [Citation(s) in RCA: 62] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2015] [Revised: 12/15/2015] [Accepted: 12/16/2015] [Indexed: 12/12/2022] Open
Abstract
High concentrations of nickel (Ni) are harmful to humans and animals. Ni targets a number of organs and produces multiple toxic effects. Apoptosis is important in Ni-induced toxicity of the kidneys, liver, nerves, and immune system. Apoptotic pathways mediated by reactive oxygen species (ROS), mitochondria, endoplasmic reticulum (ER), Fas, and c-Myc participate in Ni-induced cell apoptosis. However, the exact mechanism of apoptosis caused by Ni is still unclear. Understanding the mechanism of Ni-induced apoptosis may help in designing measures to prevent Ni toxicity.
Collapse
Affiliation(s)
- Hongrui Guo
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, China.
| | - Lian Chen
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, China.
| | - Hengmin Cui
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, China.
- College of Veterinary Medicine, Sichuan Agricultural University Ya'an, Ya'an 625014, China.
| | - Xi Peng
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, China.
- College of Veterinary Medicine, Sichuan Agricultural University Ya'an, Ya'an 625014, China.
| | - Jing Fang
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, China.
- College of Veterinary Medicine, Sichuan Agricultural University Ya'an, Ya'an 625014, China.
| | - Zhicai Zuo
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, China.
- College of Veterinary Medicine, Sichuan Agricultural University Ya'an, Ya'an 625014, China.
| | - Junliang Deng
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, China.
- College of Veterinary Medicine, Sichuan Agricultural University Ya'an, Ya'an 625014, China.
| | - Xun Wang
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, China.
- College of Veterinary Medicine, Sichuan Agricultural University Ya'an, Ya'an 625014, China.
| | - Bangyuan Wu
- Key Laboratory of Animal Diseases and Environmental Hazards of Sichuan Province, Sichuan Agricultural University, Ya'an 625014, China.
| |
Collapse
|
22
|
Muthalagu N, Junttila MR, Wiese KE, Wolf E, Morton J, Bauer B, Evan GI, Eilers M, Murphy DJ. BIM is the primary mediator of MYC-induced apoptosis in multiple solid tissues. Cell Rep 2014; 8:1347-53. [PMID: 25176652 DOI: 10.1016/j.celrep.2014.07.057] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2014] [Revised: 06/04/2014] [Accepted: 07/30/2014] [Indexed: 11/30/2022] Open
Abstract
MYC is one of the most frequently overexpressed oncogenes in human cancer, and even modestly deregulated MYC can initiate ectopic proliferation in many postmitotic cell types in vivo. Sensitization of cells to apoptosis limits MYC's oncogenic potential. However, the mechanism through which MYC induces apoptosis is controversial. Some studies implicate p19ARF-mediated stabilization of p53, followed by induction of proapoptotic BH3 proteins NOXA and PUMA, whereas others argue for direct regulation of BH3 proteins, especially BIM. Here, we use a single experimental system to systematically evaluate the roles of p19ARF and BIM during MYC-induced apoptosis, in vitro, in vivo, and in combination with a widely used chemotherapeutic, doxorubicin. We find a common specific requirement for BIM during MYC-induced apoptosis in multiple settings, which does not extend to the p53-responsive BH3 family member PUMA, and find no evidence of a role for p19ARF during MYC-induced apoptosis in the tissues examined.
Collapse
Affiliation(s)
- Nathiya Muthalagu
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Melissa R Junttila
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, 2356 Sutter Street, San Francisco, CA 94115, USA
| | - Katrin E Wiese
- Theodor Boveri Institute, Biocentre, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Elmar Wolf
- Theodor Boveri Institute, Biocentre, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Jennifer Morton
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK
| | - Barbara Bauer
- Theodor Boveri Institute, Biocentre, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Gerard I Evan
- Helen Diller Family Comprehensive Cancer Center, University of California, San Francisco, 2356 Sutter Street, San Francisco, CA 94115, USA
| | - Martin Eilers
- Theodor Boveri Institute, Biocentre, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany
| | - Daniel J Murphy
- Cancer Research UK Beatson Institute, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK; Theodor Boveri Institute, Biocentre, University of Wuerzburg, Am Hubland, 97074 Wuerzburg, Germany; Institute of Cancer Sciences, University of Glasgow, Garscube Estate, Switchback Road, Glasgow G61 1BD, UK.
| |
Collapse
|
23
|
Identifying regulatory mechanisms underlying tumorigenesis using locus expression signature analysis. Proc Natl Acad Sci U S A 2014; 111:5747-52. [PMID: 24706889 DOI: 10.1073/pnas.1309293111] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022] Open
Abstract
Retroviral insertional mutagenesis is a powerful tool for identifying putative cancer genes in mice. To uncover the regulatory mechanisms by which common insertion loci affect downstream processes, we supplemented genotyping data with genome-wide mRNA expression profiling data for 97 tumors induced by retroviral insertional mutagenesis. We developed locus expression signature analysis, an algorithm to construct and interpret the differential gene expression signature associated with each common insertion locus. Comparing locus expression signatures to promoter affinity profiles allowed us to build a detailed map of transcription factors whose protein-level regulatory activity is modulated by a particular locus. We also predicted a large set of drugs that might mitigate the effect of the insertion on tumorigenesis. Taken together, our results demonstrate the potential of a locus-specific signature approach for identifying mammalian regulatory mechanisms in a cancer context.
Collapse
|
24
|
Braun F, de Carné Trécesson S, Bertin-Ciftci J, Juin P. Protect and serve: Bcl-2 proteins as guardians and rulers of cancer cell survival. Cell Cycle 2013; 12:2937-47. [PMID: 23974114 PMCID: PMC3875667 DOI: 10.4161/cc.25972] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
It is widely accepted that anti-apoptotic Bcl-2 family members promote cancer cell survival by binding to their pro-apoptotic counterparts, thereby preventing mitochondrial outer membrane permeabilization (MOMP) and cytotoxic caspase activation. Yet, these proteins do not only function as guardians of mitochondrial permeability, preserving it, and maintaining cell survival in the face of acute or chronic stress, they also regulate non-apoptotic functions of caspases and biological processes beyond MOMP from diverse subcellular localizations and in complex with numerous binding partners outside of the Bcl-2 family. In particular, some of the non-canonical effects and functions of Bcl-2 homologs lead to an interplay with E2F-1, NFκB, and Myc transcriptional pathways, which themselves influence cancer cell growth and survival. We thus propose that, by feedback loops that we currently have only hints of, Bcl-2 proteins may act as rulers of survival signaling, predetermining the apoptotic threshold that they also directly scaffold. This underscores the robustness of the control exerted by Bcl-2 homologs over cancer cell survival, and implies that small molecules compounds currently used in the clinic to inhibit their mitochondrial activity may be not always be fully efficient to override this control.
Collapse
Affiliation(s)
- Frédérique Braun
- UMR 892 INSERM/6299 CNRS/Université de Nantes; Team 8 "Cell survival and tumor escape in breast cancer"; Institut de Recherche en Santé de l'Université de Nantes; Nantes, France
| | | | | | | |
Collapse
|
25
|
Li WD, Wu Y, Zhang L, Yan LG, Yin FZ, Ruan JS, Chen ZP, Yang GM, Yan CP, Zhao D, Lu Y, Cai BC. Characterization of xanthatin: anticancer properties and mechanisms of inhibited murine melanoma in vitro and in vivo. PHYTOMEDICINE : INTERNATIONAL JOURNAL OF PHYTOTHERAPY AND PHYTOPHARMACOLOGY 2013; 20:865-873. [PMID: 23664560 DOI: 10.1016/j.phymed.2013.03.006] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/04/2013] [Revised: 02/06/2013] [Accepted: 03/09/2013] [Indexed: 06/02/2023]
Abstract
Anti-cancer investigations on Xanthatin mainly focus on in vitro experiments. We herein reported the anti-tumor effects of Xanthatin both in vitro and in vivo. MTS assay results showed that Xanthatin had a remarkable anti-proliferative effect on B16-F10 cells. Moreover, the expression of β-catenin was up-regulated both in vitro and in vivo. Animal studies further revealed that Xanthatin killed the tumor cells around the blood vessels which contributes to reduce microvascular density extremely. All these results indicate that Xanthatin inhibited murine melanoma B16-F10 cell proliferation possibly associated with activation of Wnt/β-catenin pathway and its activity against melanoma tumor might also be relevant to inhibition of angiogenesis.
Collapse
Affiliation(s)
- Wei D Li
- Nanjing University of Chinese Medicine, Nanjing 210046, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Juin P, Geneste O, Gautier F, Depil S, Campone M. Decoding and unlocking the BCL-2 dependency of cancer cells. Nat Rev Cancer 2013; 13:455-65. [PMID: 23783119 DOI: 10.1038/nrc3538] [Citation(s) in RCA: 177] [Impact Index Per Article: 14.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Cancer cells are subject to many apoptotic stimuli that would kill them were it not for compensatory prosurvival alterations. BCL-2-like (BCL-2L) proteins contribute to such aberrant behaviour by engaging a network of interactions that is potent at promoting survival but that is also fragile: inhibition of a restricted number of interactions may suffice to trigger cancer cell death. Currently available and novel compounds that inhibit these interactions could be efficient therapeutic agents if this phenotype of BCL-2L dependence was better understood at a molecular, cellular and systems level and if it could be diagnosed by relevant biomarkers.
Collapse
Affiliation(s)
- Philippe Juin
- Team 8 Cell survival and tumor escape in breast cancer, UMR 892 INSERM / 6299 CNRS / Université de Nantes, Institut de Recherche Thérapeutique de l'Université de Nantes, 8 quai Moncousu, BP 70721, 44007 Nantes Cedex, 1 France.
| | | | | | | | | |
Collapse
|
27
|
|
28
|
Kim SH, Kang JG, Kim CS, Ihm SH, Choi MG, Yoo HJ, Lee SJ. Apigenin induces c-Myc-mediated apoptosis in FRO anaplastic thyroid carcinoma cells. Mol Cell Endocrinol 2013; 369:130-9. [PMID: 23376608 DOI: 10.1016/j.mce.2013.01.012] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/21/2012] [Revised: 01/14/2013] [Accepted: 01/17/2013] [Indexed: 01/11/2023]
Abstract
Apigenin promotes apoptosis in cancer cells. We studied the effect of apigenin on cell survival and c-Myc expression in FRO anaplastic thyroid carcinoma (ATC) cells. Apigenin caused apoptosis via the elevation of c-Myc levels in conjunction with the phosphorylation of p38 and p53. In the c-Myc siRNA-transfected and apigenin-treated cells, compared with the apigenin-treated control cells, apoptosis and phosphorylation of p38 and p53 were ameliorated. In the presence of apigenin, diminution of p38 and p53 did not affect cell survival although apigenin activated the phosphorylation of p38 and p53 via increased c-Myc levels. In conclusion, our results indicate that apigenin induces apoptosis mediated via c-Myc with concomitant phosphorylation of p53 and p38 in FRO ATC cells. These findings suggest that augmented c-Myc acts as a core regulator and is necessary for apigenin-induced apoptosis in FRO ATC cells.
Collapse
Affiliation(s)
- Si Hyoung Kim
- Division of Endocrinology and Metabolism, Department of Internal Medicine, College of Medicine, Hallym University, Chuncheon 200-704, Republic of Korea
| | | | | | | | | | | | | |
Collapse
|
29
|
Lynch JT, Somerville TDD, Spencer GJ, Huang X, Somervaille TCP. TTC5 is required to prevent apoptosis of acute myeloid leukemia stem cells. Cell Death Dis 2013; 4:e573. [PMID: 23559008 PMCID: PMC3641330 DOI: 10.1038/cddis.2013.107] [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] [Indexed: 12/27/2022]
Abstract
Using a screening strategy, we identified the tetratricopeptide repeat (TPR) motif protein, Tetratricopeptide repeat domain 5 (TTC5, also known as stress responsive activator of p300 or Strap) as required for the survival of human acute myeloid leukemia (AML) cells. TTC5 is a stress-inducible transcription cofactor known to interact directly with the histone acetyltransferase EP300 to augment the TP53 response. Knockdown (KD) of TTC5 induced apoptosis of both murine and human AML cells, with concomitant loss of clonogenic and leukemia-initiating potential; KD of EP300 elicited a similar phenotype. Consistent with the physical interaction of TTC5 and EP300, the onset of apoptosis following KD of either gene was preceded by reduced expression of BCL2 and increased expression of pro-apoptotic genes. Forced expression of BCL2 blocked apoptosis and partially rescued the clonogenic potential of AML cells following TTC5 KD. KD of both genes also led to the accumulation of MYC, an acetylation target of EP300, and the form of MYC that accumulated exhibited relative hypoacetylation at K148 and K157, residues targeted by EP300. In view of the ability of excess cellular MYC to sensitize cells to apoptosis, our data suggest a model whereby TTC5 and EP300 cooperate to prevent excessive accumulation of MYC in AML cells and their sensitization to cell death. They further reveal a hitherto unappreciated role for TTC5 in leukemic hematopoiesis.
Collapse
Affiliation(s)
- J T Lynch
- Cancer Research UK Leukaemia Biology Laboratory, Paterson Institute for Cancer Research, The University of Manchester, Manchester, UK
| | | | | | | | | |
Collapse
|
30
|
Morton JP, Sansom OJ. MYC-y mice: from tumour initiation to therapeutic targeting of endogenous MYC. Mol Oncol 2013; 7:248-58. [PMID: 23523308 PMCID: PMC5528411 DOI: 10.1016/j.molonc.2013.02.015] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2013] [Accepted: 02/18/2013] [Indexed: 12/15/2022] Open
Abstract
MYC is one of the best-studied oncogenes in terms of mouse models of malignancy. MYC overexpression has been targeted to several tissues using transgenic constructs, and more recently as mouse models have evolved, conditional systems have been developed to allow the regulation of MYC expression or activity in vivo. The ability to target MYC expression to specific tissues and cell lineages, as well as the ability to regulate that expression, has made genetically engineered mouse models (GEMM) a valuable resource for studying the importance of MYC in the process of tumourigenesis. Here we review how these models have been used to address the role of MYC in tumour initiation and maintenance, how subtle changes in levels of MYC can influence tumourigenesis, and finally the ongoing efforts to target endogenous MYC genetically and with novel therapies.
Collapse
Affiliation(s)
- Jennifer P Morton
- Beatson Institute for Cancer Research, Garscube Estate, Switchback Rd, Glasgow G61 1BD, UK
| | | |
Collapse
|
31
|
Grandori C. A high-throughput siRNA screening platform to identify MYC-synthetic lethal genes as candidate therapeutic targets. Methods Mol Biol 2013; 1012:187-200. [PMID: 24006065 DOI: 10.1007/978-1-62703-429-6_12] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Targeted therapeutics toward specific genes and pathways represent the future of oncological treatments. However, several commonly activated oncogenes, such as MYC, have proven difficult to target by pharmacological agents. To broaden the menu of potentially druggable therapeutic targets, we describe a method to detect genes essential for the survival of MYC overexpressing cells, which we will refer to as MYC-synthetic lethal genes (MYC-SL) (Toyoshima et al., Proc Natl Acad Sci USA 109:9545-9550, 2012). These genes represent candidate targets for drug development to be utilized for MYC-driven cancers as well as probes to further our understanding of the biology of MYC-driven tumorigenesis. The discovery platform includes the following components: (1) an isogenic cell system that enables overexpression of MYC without oncogene-induced senescence (OIS) response (Benanti and Galloway, Mol Cell Biol 24:2842-2852, 2004; Benanti et al., Mol Cancer Res 5:1181-1189, 2007); (2) arrayed siRNA libraries targeting individual genes; (3) automated laboratory equipment for dispensing of cells, siRNAs, and readout assays; and (4) bioinformatics and software for data mining and visualization. This flexible platform can be readily applied to other oncogenes or tumor suppressor genes.
Collapse
Affiliation(s)
- Carla Grandori
- Division of Human Biology, Fred Hutchinson Cancer Research Center, Seattle, WA, USA
| |
Collapse
|
32
|
Lu D, Littlewood TD. Methods for determining Myc-induced apoptosis. Methods Mol Biol 2013; 1012:85-98. [PMID: 24006060 DOI: 10.1007/978-1-62703-429-6_7] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
Although many oncoproteins promote cell growth and proliferation, some also possess the potential to induce cell death by apoptosis. Deregulated expression of the myc oncogene promotes apoptosis in both cultured cells and in some tissues in vivo. Here we describe techniques to detect Myc-induced apoptosis in vitro using flow cytometry and microscopy and in vivo using immunohistochemical staining.
Collapse
Affiliation(s)
- Dan Lu
- Department of Biochemistry, University of Cambridge, Cambridge, UK
| | | |
Collapse
|
33
|
Schütz AK, Hennes T, Jumpertz S, Fuchs S, Bernhagen J. Role of CSN5/JAB1 in Wnt/β-catenin activation in colorectal cancer cells. FEBS Lett 2012; 586:1645-51. [DOI: 10.1016/j.febslet.2012.04.037] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/24/2012] [Revised: 04/12/2012] [Accepted: 04/18/2012] [Indexed: 11/16/2022]
|
34
|
Järvinen K, Hotti A, Santos L, Nummela P, Hölttä E. Caspase-8, c-FLIP, and caspase-9 in c-Myc-induced apoptosis of fibroblasts. Exp Cell Res 2011; 317:2602-15. [DOI: 10.1016/j.yexcr.2011.08.014] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/05/2011] [Revised: 08/10/2011] [Accepted: 08/22/2011] [Indexed: 01/02/2023]
|
35
|
Alam S, Bowser BS, Conway MJ, Israr M, Tandon A, Meyers C. Adeno-associated virus type 2 infection activates caspase dependent and independent apoptosis in multiple breast cancer lines but not in normal mammary epithelial cells. Mol Cancer 2011; 10:97. [PMID: 21827643 PMCID: PMC3199901 DOI: 10.1186/1476-4598-10-97] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2011] [Accepted: 08/09/2011] [Indexed: 01/28/2023] Open
Abstract
Background In normal cells proliferation and apoptosis are tightly regulated, whereas in tumor cells the balance is shifted in favor of increased proliferation and reduced apoptosis. Anticancer agents mediate tumor cell death via targeting multiple pathways of programmed cell death. We have reported that the non-pathogenic, tumor suppressive Adeno-Associated Virus Type 2 (AAV2) induces apoptosis in Human Papillomavirus (HPV) positive cervical cancer cells, but not in normal keratinocytes. In the current study, we examined the potential of AAV2 to inhibit proliferation of MCF-7 and MDA-MB-468 (both weakly invasive), as well as MDA-MB-231 (highly invasive) human breast cancer derived cell lines. As controls, we used normal human mammary epithelial cells (nHMECs) isolated from tissue biopsies of patients undergoing breast reduction surgery. Results AAV2 infected MCF-7 line underwent caspase-independent, and MDA-MB-468 and MDA-MB-231 cell lines underwent caspase-dependent apoptosis. Death of MDA-MB-468 cells was marked by caspase-9 activation, whereas death of MDA-MB-231 cells was marked by activation of both caspase-8 and caspase-9, and resembled a mixture of apoptotic and necrotic cell death. Cellular demise was correlated with the ability of AAV2 to productively infect and differentially express AAV2 non-structural proteins: Rep78, Rep68 and Rep40, dependent on the cell line. Cell death in the MCF-7 and MDA-MB-231 lines coincided with increased S phase entry, whereas the MDA-MB-468 cells increasingly entered into G2. AAV2 infection led to decreased cell viability which correlated with increased expression of proliferation markers c-Myc and Ki-67. In contrast, nHMECs that were infected with AAV2 failed to establish productive infection or undergo apoptosis. Conclusion AAV2 regulated enrichment of cell cycle check-point functions in G1/S, S and G2 phases could create a favorable environment for Rep protein expression. Inherent Rep associated endonuclease activity and AAV2 genomic hair-pin ends have the potential to induce a cellular DNA damage response, which could act in tandem with c-Myc regulated/sensitized apoptosis induction. In contrast, failure of AAV2 to productively infect nHMECs could be clinically advantageous. Identifying the molecular mechanisms of AAV2 targeted cell cycle regulation of death inducing signals could be harnessed for developing novel therapeutics for weakly invasive as well as aggressive breast cancer types.
Collapse
Affiliation(s)
- Samina Alam
- Department of Microbiology and Immunology, Pennsylvania State University College of Medicine, 500 University Drive, Hershey, PA 17033, USA
| | | | | | | | | | | |
Collapse
|
36
|
Allen TD, Zhu CQ, Jones KD, Yanagawa N, Tsao MS, Bishop JM. Interaction between MYC and MCL1 in the genesis and outcome of non-small-cell lung cancer. Cancer Res 2011; 71:2212-21. [PMID: 21406400 DOI: 10.1158/0008-5472.can-10-3590] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
MYC exerts both positive and negative functions in cancer cells, such that its procancerous effects are unmasked only after its anticancer effects are blocked. Here we used multiple mouse models of lung adenocarcinoma to identify genetic events that can cooperate with MYC activation to promote the genesis of non-small-cell lung cancer (NSCLC), the most common form of lung cancer in humans. MYC overexpression targeted to pulmonary alveolar cells was sufficient to induce lung adenomas and carcinomas. Tumorigenesis was assisted by either spontaneous mutations in Kras or experimental introduction of activated RAS, but investigations revealed that additional events were required to circumvent apoptosis, one of the most significant negative functions exerted by MYC. We determined that overexpression of the antiapoptotic protein MCL1 was sufficient to circumvent apoptosis in this setting. Previous clinical studies have indicated that prognosis of human NSCLC is not associated with MCL1, despite its overexpression in many NSCLCs. In reexamining the prognostic value in this setting, we found that MCL1 overexpression does correlate with poor patient survival, but only when accompanied by MYC overexpression. Our findings therefore produce a convergence of mouse and human results that explain how MCL1 can block an important negative consequence of MYC overexpression in both experimental models and clinical cases of NSCLC.
Collapse
Affiliation(s)
- Thaddeus D Allen
- G.W. Hooper Research Foundation, University of California, San Francisco, USA.
| | | | | | | | | | | |
Collapse
|
37
|
Zhang T, Tang SS, Jin X, Liu FY, Zhang CM, Zhao WX, Zhang S, Sun CD, Xiao XL. c-Myc influences olaquindox-induced apoptosis in human hepatoma G2 cells. Mol Cell Biochem 2011; 354:253-61. [DOI: 10.1007/s11010-011-0825-2] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2010] [Accepted: 04/15/2011] [Indexed: 12/27/2022]
|
38
|
Zhang L, Bao Y, Li J. Nuclear respiratory factor-1 is involved in mitochondrial dysfunction induced by benzo(a)pyrene in human bronchial epithelial cells. Basic Clin Pharmacol Toxicol 2011; 109:115-22. [PMID: 21401886 DOI: 10.1111/j.1742-7843.2011.00697.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
Abstract
In this study, we investigated the role of nuclear respiratory factor-1(NRF-1) in benzo(a)pyrene (BaP)-induced mitochondrial events in human bronchial epithelial cells (16HBE). Cytotoxicity was determined with MTT assay, and apoptosis was measured by flow cytometry. The results showed that BaP inhibited cell proliferation in a dose-dependent manner and induced apoptosis in 16HBE cells. Time-dependent reactive oxygen species (ROS) generation induced by BaP was observed in 16HBE cells. The loss of mitochondrial membrane permeability transition (MPT) was obtained by a laser scanning confocal microscope, and the decreasing ATP level was detected by a Cell-Titer-Glo(®) Luminescent Cell Viability Assay. Results of western blotting assay revealed that both NRF-1 and mitochondrial transcription factor A (mtTFA) decreased in 12-μM BaP-treated cells at both 12 and 24 hr. The results of RT-PCR indicate that NRF-1 and mtTFA mRNA in 16HBE cells were not changed after BaP treatment 12 or 24 hr. Down-regulation of NRF-1 by shRNA further reduced the loss of MPT and increased ROS generation in response to BaP treatment. Therefore, our results demonstrate that NRF-1 is responsible for BaP-induced mitochondrial dysfunction in 16HBE cells and associated with the level of mtTFA protein, loss of MPT and ROS overproduction.
Collapse
Affiliation(s)
- Lijuan Zhang
- Department of Prevention, Tongji University School of Medicine, Shanghai, China
| | | | | |
Collapse
|
39
|
Lai Y, Qiao M, Song M, Weintraub ST, Shiio Y. Quantitative proteomics identifies the Myb-binding protein p160 as a novel target of the von Hippel-Lindau tumor suppressor. PLoS One 2011; 6:e16975. [PMID: 21386990 PMCID: PMC3046137 DOI: 10.1371/journal.pone.0016975] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/26/2010] [Accepted: 01/11/2011] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND The von Hippel-Lindau (VHL) tumor suppressor gene encodes a component of a ubiquitin ligase complex, which is best understood as a negative regulator of hypoxia inducible factor (HIF). VHL ubiquitinates and degrades the α subunits of HIF, and this is proposed to suppress tumorigenesis and tumor angiogenesis. However, several lines of evidence suggest that there are unidentified substrates or targets for VHL that play important roles in tumor suppression. METHODOLOGY/PRINCIPAL FINDINGS Employing quantitative proteomics, we developed an approach to systematically identify the substrates of ubiquitin ligases and using this method, we identified the Myb-binding protein p160 as a novel substrate of VHL. CONCLUSIONS/SIGNIFICANCE A major barrier to understanding the functions of ubiquitin ligases has been the difficulty in pinpointing their ubiquitination substrates. The quantitative proteomics approach we devised for the identification of VHL substrates will be widely applicable to other ubiquitin ligases.
Collapse
Affiliation(s)
- Yanlai Lai
- Greehey Children's Cancer Research Institute, San Antonio, Texas, United States of America
| | - Mei Qiao
- Greehey Children's Cancer Research Institute, San Antonio, Texas, United States of America
| | - Meihua Song
- Greehey Children's Cancer Research Institute, San Antonio, Texas, United States of America
| | - Susan T. Weintraub
- Department of Biochemistry, The University of Texas Health Science Center, San Antonio, Texas, United States of America
| | - Yuzuru Shiio
- Greehey Children's Cancer Research Institute, San Antonio, Texas, United States of America
- Department of Biochemistry, The University of Texas Health Science Center, San Antonio, Texas, United States of America
| |
Collapse
|
40
|
Gravina GL, Festuccia C, Marampon F, Popov VM, Pestell RG, Zani BM, Tombolini V. Biological rationale for the use of DNA methyltransferase inhibitors as new strategy for modulation of tumor response to chemotherapy and radiation. Mol Cancer 2010; 9:305. [PMID: 21108789 PMCID: PMC3001713 DOI: 10.1186/1476-4598-9-305] [Citation(s) in RCA: 89] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/18/2010] [Accepted: 11/25/2010] [Indexed: 12/31/2022] Open
Abstract
Epigenetic modifications play a key role in the patho-physiology of many tumors and the current use of agents targeting epigenetic changes has become a topic of intense interest in cancer research. DNA methyltransferase (DNMT) inhibitors represent a promising class of epigenetic modulators. Research performed yielded promising anti-tumorigenic activity for these agents in vitro and in vivo against a variety of hematologic and solid tumors. These epigenetic modulators cause cell cycle and growth arrest, differentiation and apoptosis. Rationale for combining these agents with cytotoxic therapy or radiation is straightforward since the use of DNMT inhibitor offers greatly improved access for cytotoxic agents or radiation for targeting DNA-protein complex. The positive results obtained with these combined approaches in preclinical cancer models demonstrate the potential impact DNMT inhibitors may have in treatments of different cancer types. Therefore, as the emerging interest in use of DNMT inhibitors as a potential chemo- or radiation sensitizers is constantly increasing, further clinical investigations are inevitable in order to finalize and confirm the consistency of current observations.The present article will provide a brief review of the biological significance and rationale for the clinical potential of DNMT inhibitors in combination with other chemotherapeutics or ionizing radiation. The molecular basis and mechanisms of action for these combined treatments will be discussed herein.
Collapse
Affiliation(s)
- Giovanni L Gravina
- Department of Experimental Medicine, Division of Radiation Oncology, S, Salvatore Hospital, L'Aquila, University of L'Aquila, Medical School, L'Aquila 67100, Italy.
| | | | | | | | | | | | | |
Collapse
|
41
|
N,N-Dimethyl phytosphingosine sensitizes HL-60/MX2, a multidrug-resistant variant of HL-60 cells, to doxorubicin-induced cytotoxicity through ROS-mediated release of cytochrome c and AIF. Apoptosis 2010; 15:982-93. [DOI: 10.1007/s10495-010-0512-x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
|
42
|
Albihn A, Johnsen JI, Henriksson MA. MYC in oncogenesis and as a target for cancer therapies. Adv Cancer Res 2010; 107:163-224. [PMID: 20399964 DOI: 10.1016/s0065-230x(10)07006-5] [Citation(s) in RCA: 186] [Impact Index Per Article: 12.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/19/2022]
Abstract
MYC proteins (c-MYC, MYCN, and MYCL) regulate processes involved in many if not all aspects of cell fate. Therefore, it is not surprising that the MYC genes are deregulated in several human neoplasias as a result from genetic and epigenetic alterations. The near "omnipotency" together with the many levels of regulation makes MYC an attractive target for tumor intervention therapy. Here, we summarize some of the current understanding of MYC function and provide an overview of different cancer forms with MYC deregulation. We also describe available treatments and highlight novel approaches in the pursuit for MYC-targeting therapies. These efforts, at different stages of development, constitute a promising platform for novel, more specific treatments with fewer side effects. If successful a MYC-targeting therapy has the potential for tailored treatment of a large number of different tumors.
Collapse
Affiliation(s)
- Ami Albihn
- Department of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | | | | |
Collapse
|
43
|
Jaagsiekte sheep retrovirus transformation in Madin-Darby canine kidney epithelial cell three-dimensional culture. J Virol 2010; 84:5379-90. [PMID: 20219922 DOI: 10.1128/jvi.02323-09] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/28/2023] Open
Abstract
Jaagsiekte sheep retrovirus (JSRV) is the causative agent of a contagious lung cancer in sheep that shares similarities with human bronchioloalveolar carcinoma (BAC). JSRV is unique because the envelope gene (env) is the oncogene, as it can transform cells in culture and induce tumors in animals. The phosphatidylinositol 3-kinase (PI3K)-Akt-mTOR and H/N-Ras-MEK-mitogen-activated protein kinase (MAPK) pathways have been shown to be critical for Env transformation. However, the question still remains of how disruption of these pathways relates to tumor formation. To address this, JSRV Env transformation was studied in the context of epithelial structure, using the polarized Madin-Darby canine kidney (MDCK) epithelial cell three-dimensional (3-D) culture system. The results indicated that JSRV Env-transformed MDCK cells were larger and had full or multiple lumens, in contrast to the single lumens observed in controls. The altered phenotype was largely mediated by an increase in proliferation, in addition to overcoming the proliferative suppression signal. JSRV Env was not found to disrupt polarity or tight junctions or to inhibit lumen apoptosis. The PI3K-Akt-mTOR pathway was important for Env transformation in MDCK cells, although the mechanisms of action differed in 3-D and monolayer cultures. PI3K-dependent signaling to mTOR occurred in monolayers, while PI3K-independent signaling to mTOR occurred in 3-D culture. In contrast, the H/N-Ras-MEK-MAPK pathway was found to be inhibitory to transformation in both normal and transformed MDCK cells in 3-D culture. However, in monolayer culture, inhibition of MEK reverted the transformed phenotype, suggesting a different mechanism(s) of action in monolayer versus 3-D culture.
Collapse
|
44
|
Modulation of cellular migration and survival by c-Myc through the downregulation of urokinase (uPA) and uPA receptor. Mol Cell Biol 2010; 30:1838-51. [PMID: 20123981 DOI: 10.1128/mcb.01442-09] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
It has been proposed that c-Myc proapoptotic activity accounts for most of its restraint of tumor formation. We established a telomerase-immortalized human epithelial cell line expressing an activatable c-Myc protein. We found that c-Myc activation induces, in addition to increased sensitivity to apoptosis, reductions in cell motility and invasiveness. Transcriptome analysis revealed that urokinase (uPA) and uPA receptor (uPAR) were strongly downregulated by c-Myc. Evidence is provided that the repression of uPA and uPAR may account for most of the antimigratory and proapoptotic activities of c-Myc. c-Myc is known to cooperate with Ras in cellular transformation. We therefore investigated if this cooperation could converge in the control of uPA/uPAR expression. We found that Ras is able to block the effects of c-Myc activation on apoptosis and cellular motility but not on cell invasiveness. Accordingly, the activation of c-Myc in the context of Ras expression had only minor influence on uPAR expression but still had a profound repressive effect on uPA expression. Thus, the differential regulation of uPA and uPAR by c-Myc and Ras correlates with the effects of these two oncoproteins on cell motility, invasiveness, and survival. In conclusion, we have discovered a novel link between c-Myc and uPA/uPAR. We propose that reductions of cell motility and invasiveness could contribute to the inhibition of tumorigenesis by c-Myc and that the regulation of uPA and uPAR expression may be a component of the ability of c-Myc to reduce motility and invasiveness.
Collapse
|
45
|
Guerra L, Albihn A, Tronnersjö S, Yan Q, Guidi R, Stenerlöw B, Sterzenbach T, Josenhans C, Fox JG, Schauer DB, Thelestam M, Larsson LG, Henriksson M, Frisan T. Myc is required for activation of the ATM-dependent checkpoints in response to DNA damage. PLoS One 2010; 5:e8924. [PMID: 20111719 PMCID: PMC2811743 DOI: 10.1371/journal.pone.0008924] [Citation(s) in RCA: 57] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2009] [Accepted: 01/05/2010] [Indexed: 01/20/2023] Open
Abstract
Background The MYC protein controls cellular functions such as differentiation, proliferation, and apoptosis. In response to genotoxic agents, cells overexpressing MYC undergo apoptosis. However, the MYC-regulated effectors acting upstream of the mitochondrial apoptotic pathway are still unknown. Principal Findings In this study, we demonstrate that expression of Myc is required to activate the Ataxia telangiectasia mutated (ATM)-dependent DNA damage checkpoint responses in rat cell lines exposed to ionizing radiation (IR) or the bacterial cytolethal distending toxin (CDT). Phosphorylation of the ATM kinase and its downstream effectors, such as histone H2AX, were impaired in the myc null cell line HO15.19, compared to the myc positive TGR-1 and HOmyc3 cells. Nuclear foci formation of the Nijmegen Breakage Syndrome (Nbs) 1 protein, essential for efficient ATM activation, was also reduced in absence of myc. Knock down of the endogenous levels of MYC by siRNA in the human cell line HCT116 resulted in decreased ATM and CHK2 phosphorylation in response to irradiation. Conversely, cell death induced by UV irradiation, known to activate the ATR-dependent checkpoint, was similar in all the cell lines, independently of the myc status. Conclusion These data demonstrate that MYC contributes to the activation of the ATM-dependent checkpoint responses, leading to cell death in response to specific genotoxic stimuli.
Collapse
Affiliation(s)
- Lina Guerra
- Departments of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Ami Albihn
- Departments of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Susanna Tronnersjö
- Departments of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Qinzi Yan
- Departments of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Riccardo Guidi
- Departments of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Bo Stenerlöw
- Division of Biomedical Radiation Sciences, Rudbeck Laboratory, Uppsala University, Uppsala, Sweden
| | - Torsten Sterzenbach
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - Christine Josenhans
- Institute for Medical Microbiology and Hospital Epidemiology, Hannover Medical School, Hannover, Germany
| | - James G. Fox
- Department of Biological Engineering, Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - David B. Schauer
- Department of Biological Engineering, Division of Comparative Medicine, Massachusetts Institute of Technology, Cambridge, Massachusetts, United States of America
| | - Monica Thelestam
- Departments of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
| | - Lars-Gunnar Larsson
- Departments of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
- Department of Plant Biology and Forest Genetics, Swedish University of Agricultural Sciences, Uppsala, Sweden
| | - Marie Henriksson
- Departments of Microbiology, Tumor and Cell Biology, Karolinska Institutet, Stockholm, Sweden
| | - Teresa Frisan
- Departments of Cell and Molecular Biology, Karolinska Institutet, Stockholm, Sweden
- * E-mail:
| |
Collapse
|
46
|
Tsiftsoglou AS, Vizirianakis IS, Strouboulis J. Erythropoiesis: model systems, molecular regulators, and developmental programs. IUBMB Life 2009; 61:800-30. [PMID: 19621348 DOI: 10.1002/iub.226] [Citation(s) in RCA: 157] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Human erythropoiesis is a complex multistep developmental process that begins at the level of pluripotent hematopoietic stem cells (HSCs) at bone marrow microenvironment (HSCs niche) and terminates with the production of erythrocytes (RBCs). This review covers the basic and contemporary aspects of erythropoiesis. These include the: (a) cell-lineage restricted pathways of differentiation originated from HSCs and going downward toward the blood cell development; (b) model systems employed to study erythropoiesis in culture (erythroleukemia cell lines and embryonic stem cells) and in vivo (knockout animals: avian, mice, zebrafish, and xenopus); (c) key regulators of erythropoiesis (iron, hypoxia, stress, and growth factors); (d) signaling pathways operating at hematopoietic stem cell niche for homeostatic regulation of self renewal (SCF/c-kit receptor, Wnt, Notch, and Hox) and for erythroid differentiation (HIF and EpoR). Furthermore, this review presents the mechanisms through which transcriptional factors (GATA-1, FOG-1, TAL-1/SCL/MO2/Ldb1/E2A, EKLF, Gfi-1b, and BCL11A) and miRNAs regulate gene pattern expression during erythroid differentiation. New insights regarding the transcriptional regulation of alpha- and beta-globin gene clusters were also presented. Emphasis was also given on (i) the developmental program of erythropoiesis, which consists of commitment to terminal erythroid maturation and hemoglobin production, (two closely coordinated events of erythropoieis) and (ii) the capacity of human embryonic and umbilical cord blood (UCB) stem cells to differentiate and produce RBCs in culture with highly selective media. These most recent developments will eventually permit customized red blood cell production needed for transfusion.
Collapse
Affiliation(s)
- Asterios S Tsiftsoglou
- Laboratory of Pharmacology, Department of Pharmaceutical Sciences, Aristotle University of Thessaloniki, Thessaloniki, Greece.
| | | | | |
Collapse
|
47
|
Gatti G, Maresca G, Natoli M, Florenzano F, Nicolin A, Felsani A, D'Agnano I. MYC prevents apoptosis and enhances endoreduplication induced by paclitaxel. PLoS One 2009; 4:e5442. [PMID: 19421315 PMCID: PMC2673584 DOI: 10.1371/journal.pone.0005442] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2008] [Accepted: 03/22/2009] [Indexed: 01/02/2023] Open
Abstract
Background The role of the MYC oncogene in the apoptotic pathways is not fully understood. MYC has been reported to protect cells from apoptosis activation but also to sensitize cells to apoptotic stimuli. We have previously demonstrated that the down-regulation of Myc protein activates apoptosis in melanoma cells and increases the susceptibility of cells to various antitumoral treatments. Beyond the well-known role in the G1→S transition, MYC is also involved in the G2-M cell cycle phases regulation. Methodology/Principal Findings In this study we have investigated how MYC could influence cell survival signalling during G2 and M phases. We used the microtubules damaging agent paclitaxel (PTX), to arrest the cells in the M phase, in a p53 mutated melanoma cell line with modulated Myc level and activity. An overexpression of Myc protein is able to increase endoreduplication favoring the survival of cells exposed to antimitotic poisoning. The PTX-induced endoreduplication is associated in Myc overexpressing cells with a reduced expression of MAD2, essential component of the molecular core of the spindle assembly checkpoint (SAC), indicating an impairment of this checkpoint. In addition, for the first time we have localized Myc protein at the spindle poles (centrosomes) during pro-metaphase in different cell lines. Conclusions The presence of Myc at the poles during the prometaphase could be necessary for the Myc-mediated attenuation of the SAC and the subsequent induction of endoreduplication. In addition, our data strongly suggest that the use of taxane in antitumor therapeutic strategies should be rationally based on the molecular profile of the individual tumor by specifically analyzing Myc expression levels.
Collapse
Affiliation(s)
- Giuliana Gatti
- Department of Pharmacology, University of Milan, Milan, Italy
| | - Giovanna Maresca
- CNR, Institute of Neurobiology and Molecular Medicine, European Brain Research Institute, S. Lucia Foundation, Rome, Italy
| | - Manuela Natoli
- CNR, Institute of Neurobiology and Molecular Medicine, European Brain Research Institute, S. Lucia Foundation, Rome, Italy
| | - Fulvio Florenzano
- CNR, Institute of Neurobiology and Molecular Medicine, European Brain Research Institute, S. Lucia Foundation, Rome, Italy
| | - Angelo Nicolin
- Department of Pharmacology, University of Milan, Milan, Italy
| | - Armando Felsani
- CNR, Institute of Neurobiology and Molecular Medicine, European Brain Research Institute, S. Lucia Foundation, Rome, Italy
- Genomnia, Milan, Italy
- * E-mail: (AF); (ID)
| | - Igea D'Agnano
- CNR, Institute of Neurobiology and Molecular Medicine, European Brain Research Institute, S. Lucia Foundation, Rome, Italy
- * E-mail: (AF); (ID)
| |
Collapse
|
48
|
Radziszewska A, Schroer SA, Choi D, Tajmir P, Radulovich N, Ho JC, Wang L, Liadis N, Hakem R, Tsao MS, Penn LZ, Evan GI, Woo M. Absence of caspase-3 protects pancreatic {beta}-cells from c-Myc-induced apoptosis without leading to tumor formation. J Biol Chem 2009; 284:10947-56. [PMID: 19213729 DOI: 10.1074/jbc.m806960200] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
c-Myc is a powerful trigger of beta-cell apoptosis, proliferation, and dedifferentiation in rodent islets in vivo. In a transgenic mouse model, c-Myc induction causes rapid beta-cell apoptosis and overt diabetes. When suppression of apoptosis is achieved by overexpression of Bcl-x(L) in an inducible model of c-Myc activation, a full spectrum of tumor development, including distant metastasis, occurs. Caspase-3 is a key pro-apoptotic protein involved in the execution phase of multiple apoptotic pathways. To test whether caspase-3 is an essential mediator of apoptosis in this model of tumorigenesis, we generated caspase-3 knock-out mice containing the inducible c-myc transgene (c-Myc(+)Casp3(-/-)). In contrast to Bcl-x(L)-overexpressing c-Myc(+) mice, c-Myc(+)Casp3(-/-) mice remained euglycemic for up to 30 days of c-Myc activation, and there was no evidence of tumor formation. Interestingly, caspase-3 deletion also led to the suppression of proliferation, perhaps through regulation of the cell cycle inhibitory protein p27, suggesting a possible mechanism for maintaining a balance between suppression of apoptosis and excessive proliferation in the context of c-Myc activation. Additionally, c-Myc-activated Casp3(-/-) mice were protected from streptozotocin-induced diabetes. Our studies demonstrate that caspase-3 deletion confers protection from c-Myc-induced apoptosis and diabetes development without unwanted tumorigenic effects. These results may lead to further elucidation of the mechanisms of c-Myc biology relevant to beta-cells, which may result in novel therapeutic strategies for diabetes.
Collapse
Affiliation(s)
- Anna Radziszewska
- Department of Medical Biophysics and Institute of Medical Science, Ontario Cancer Institute, University of Toronto, Toronto, Ontario M5G 2M9, Canada
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
49
|
Amendola D, De Salvo M, Marchese R, Verga Falzacappa C, Stigliano A, Carico E, Brunetti E, Moscarini M, Bucci B. Myc down-regulation affects cyclin D1/cdk4 activity and induces apoptosis via Smac/Diablo pathway in an astrocytoma cell line. Cell Prolif 2009; 42:94-109. [PMID: 19143767 DOI: 10.1111/j.1365-2184.2008.00576.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
OBJECTIVES We investigated the antiproliferative effect of Myc down-regulation via cell proliferation inhibition, cell cycle perturbation and apoptosis in two human astrocytoma models (T98G and ADF) steadily expressing an inducible c-myc Anti-sense RNA. MATERIALS AND METHODS Cell growth experiments were performed using the trypan blue dye exclusion test and cell cycle analysis was evaluated by flow cytometry. Cell cycle molecules were detected by Western blot analysis, co-immunoprecipitation and reverse transcription-polymerase chain reaction assays. RESULTS We showed that Myc down-regulation in astrocytoma cells led to G1 accumulation and an inhibition of cell proliferation characterized by S phase delay. Co-immunoprecipitation experiments detected formation of inactive cyclin D1/cdk4 complexes as evaluated by presence of an active unphosphorylated form of retinoblastoma protein, the best characterized target substrate for cyclin D1/cdk4 complex, in ADF pINDc-myc anti-sense 7 cells. We also found that either p57Kip2 "apice" or p27Kip1 "apice" inhibitors bound to cyclin D1/cdk4 complex, thus, suggesting that they cooperated to inhibit the activity of cyclin D1/cdk4. Moreover, c-Myc down-regulation led to activation of the apoptotic mitochondrial pathway, characterized by release of cytochrome c and Smac/Diablo proteins and by reduction of c-IAP levels through activation of proteasome-mediated protein degradation system. CONCLUSIONS Our results suggest that c-Myc could be considered as a good target for the study of new approaches in anticancer astrocytoma treatment.
Collapse
Affiliation(s)
- D Amendola
- Centro Ricerca S. Pietro, Fatebenefratelli Hospital, Rome, Italy
| | | | | | | | | | | | | | | | | |
Collapse
|
50
|
Meola J, Rosa e Silva JC, Dentillo DB, da Silva WA, Veiga-Castelli LC, Bernardes LADS, Ferriani RA, de Paz CCP, Giuliatti S, Martelli L. Differentially expressed genes in eutopic and ectopic endometrium of women with endometriosis. Fertil Steril 2009; 93:1750-73. [PMID: 19200988 DOI: 10.1016/j.fertnstert.2008.12.058] [Citation(s) in RCA: 79] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/13/2008] [Revised: 12/10/2008] [Accepted: 12/12/2008] [Indexed: 10/21/2022]
Abstract
OBJECTIVE To elucidate the potential mechanisms involved in the physiopathology of endometriosis. We analyzed the differential gene expression profiles of eutopic and ectopic tissues from women with endometriosis. DESIGN Prospective laboratory study. SETTING University hospital. PATIENT(S) Seventeen patients in whom endometriosis was diagnosed and 11 healthy fertile women. INTERVENTION(S) Endometrial biopsy specimens from the endometrium of healthy women without endometriosis and from the eutopic and ectopic endometrium tissues of patients with endometriosis were obtained in the early proliferative phase of the menstrual cycle. MAIN OUTCOME MEASURE(S) Six paired samples of eutopic and ectopic tissue were analyzed by subtractive hybridization. To evaluate the expression of genes found by rapid subtraction hybridization methods, we measured CTGF, SPARC, MYC, MMP, and IGFBP1 genes by real-time polymerase chain reaction in all samples. RESULT(S) This study identified 291 deregulated genes in the endometriotic lesions. Significant expression differences were obtained for SPARC, MYC, and IGFBP1 in the peritoneal lesions and for MMP3 in the ovarian endometriomas. Additionally, significant differences were obtained for SPARC and IGFBP1 between the peritoneal and ovarian lesions. No significant differences were found for the studied genes between the control and the eutopic endometrium. CONCLUSION(S) This study identified 291 genes with differential expression in endometriotic lesions. The deregulation of the SPARC, MYC, MMP3, and IGFBPI genes may be responsible for the loss of cellular homeostasis in endometriotic lesions.
Collapse
Affiliation(s)
- Juliana Meola
- Department of Genetics, School of Medicine of Ribeirão Preto, University of São Paulo, Ribeirão Preto, São Paulo, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|