1
|
Ullah MA, Moin AT, Nipa JF, Islam NN, Johora FT, Chowdhury RH, Islam S. Exploring risk factors and molecular targets in leukemia patients with COVID-19: a bioinformatics analysis of differential gene expression. J Leukoc Biol 2024; 115:723-737. [PMID: 38323674 DOI: 10.1093/jleuko/qiae002] [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: 09/24/2023] [Revised: 11/13/2023] [Accepted: 12/14/2023] [Indexed: 02/08/2024] Open
Abstract
The molecular mechanism of COVID-19's pathogenic effects in leukemia patients is still poorly known. Our study investigated the possible disease mechanism of COVID-19 and its associated risk factors in patients with leukemia utilizing differential gene expression analysis. We also employed network-based approaches to identify molecular targets that could potentially diagnose and treat COVID-19-infected leukemia patients. Our study demonstrated a shared set of 60 genes that are expressed differentially among patients with leukemia and COVID-19. Most of these genes are expressed in blood and bone marrow tissues and are predominantly implicated in the pathogenesis of different hematologic malignancies, increasingly imperiling COVID-19 morbidity and mortality among the affected patients. Additionally, we also found that COVID-19 may influence the expression of several cancer-associated genes in leukemia patients, such as CCR7, LEF1, and 13 candidate cancer-driver genes. Furthermore, our findings reveal that COVID-19 may predispose leukemia patients to altered blood homeostasis, increase the risk of COVID-19-related liver injury, and deteriorate leukemia-associated injury and patient prognosis. Our findings imply that molecular signatures, like transcription factors, proteins such as TOP21, and 25 different microRNAs, may be potential targets for diagnosing and treating COVID-19-infected leukemia patients. Nevertheless, additional experimental studies will contribute to further validating the study's findings.
Collapse
Affiliation(s)
- Md Asad Ullah
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| | - Abu Tayab Moin
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Hathazari, Chattogram-4331, Bangladesh
| | - Jannatul Ferdous Nipa
- Department of Genetic Engineering and Biotechnology, East West University, Aftabnagar, Dhaka-1212, Bangladesh
| | - Nafisa Nawal Islam
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| | - Fatema Tuz Johora
- Department of Biotechnology and Genetic Engineering, Faculty of Biological Sciences, Jahangirnagar University, Savar, Dhaka-1342, Bangladesh
| | - Rahee Hasan Chowdhury
- Department of Genetic Engineering and Biotechnology, Faculty of Biological Sciences, University of Chittagong, Hathazari, Chattogram-4331, Bangladesh
| | - Saiful Islam
- Bangladesh Council of Scientific and Industrial Research (BCSIR), Chattogram Laboratories, Chittagong Cantonment, Chattogram-4220, Bangladesh
| |
Collapse
|
2
|
Shen Y, Ren H, Davshilt T, Tian S, Wang X, Yi M, Ulaangerel T, Li B, Dugarjav M, Bou G. The transcriptome landscapes of allantochorion and vitelline-chorion in equine day 30 conceptus. Front Cell Dev Biol 2022; 10:958205. [PMID: 35990610 PMCID: PMC9386053 DOI: 10.3389/fcell.2022.958205] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2022] [Accepted: 07/11/2022] [Indexed: 11/13/2022] Open
Abstract
During equine early gestation, trophectoderm forms chorion tissue, which is composed of two parts that one is covering allantoin, called allantochorion (AC) and another is covering yolk sac, which here we call vitelline-chorion (VC). Given that little is known about the equine trophoblast-derived chorion differentiation at an early stage, we first compared the transcriptome of AC and VC of day 30 equine conceptus based on RNA-sequencing. As a result, we found that compared to VC, there are 484 DEGs, including 305 up- and 179 down-regulated genes in AC. GO and KEGG analysis indicated that up-regulated genes in AC are mainly cell proliferation and cell adhesion-related genes, participating in allantois expansion and allantochorionic-placenta formation; dominant genes in VC are extracellular exosome and other cell adhesion-related genes implicated in direct and indirect conceptus-maternal communication. Additionally, as for the progenitor chorion tissue of equine chorionic gonadotropin secreting endometrium cup-the chorionic girdle (CG), which locates at the junction of the dilating AC and regressing VC, we revealed its unique gene expression pattern and the gene regulation during its further differentiation in vitro. Collectively, this study sheds light on the molecular events regarding the trophoblast differentiation and function at an early stage of the equine preimplantation conceptus.
Collapse
Affiliation(s)
| | | | | | | | | | | | | | | | - Manglai Dugarjav
- College of Animal Science, Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Inner Mongolia Agricultural University, Hohhot, China
| | - Gerelchimeg Bou
- College of Animal Science, Inner Mongolia Key Laboratory of Equine Genetics, Breeding and Reproduction, Inner Mongolia Agricultural University, Hohhot, China
| |
Collapse
|
3
|
Toll-like Receptor 4, Osteoblasts and Leukemogenesis; the Lesson from Acute Myeloid Leukemia. MOLECULES (BASEL, SWITZERLAND) 2022; 27:molecules27030735. [PMID: 35163998 PMCID: PMC8838156 DOI: 10.3390/molecules27030735] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 11/10/2021] [Revised: 01/13/2022] [Accepted: 01/19/2022] [Indexed: 12/29/2022]
Abstract
Toll-like receptor 4 (TLR4) is a pattern-recognizing receptor that can bind exogenous and endogenous ligands. It is expressed by acute myeloid leukemia (AML) cells, several bone marrow stromal cells, and nonleukemic cells involved in inflammation. TLR4 can bind a wide range of endogenous ligands that are present in the bone marrow microenvironment. Furthermore, the TLR4-expressing nonleukemic bone marrow cells include various mesenchymal cells, endothelial cells, differentiated myeloid cells, and inflammatory/immunocompetent cells. Osteoblasts are important stem cell supporting cells localized to the stem cell niches, and they support the proliferation and survival of primary AML cells. These supporting effects are mediated by the bidirectional crosstalk between AML cells and supportive osteoblasts through the local cytokine network. Finally, TLR4 is also important for the defense against complicating infections in neutropenic patients, and it seems to be involved in the regulation of inflammatory and immunological reactions in patients treated with allogeneic stem cell transplantation. Thus, TLR4 has direct effects on primary AML cells, and it has indirect effects on the leukemic cells through modulation of their supporting neighboring bone marrow stromal cells (i.e., modulation of stem cell niches, regulation of angiogenesis). Furthermore, in allotransplant recipients TLR4 can modulate inflammatory and potentially antileukemic immune reactivity. The use of TLR4 targeting as an antileukemic treatment will therefore depend both on the biology of the AML cells, the biological context of the AML cells, aging effects reflected both in the AML and the stromal cells and the additional antileukemic treatment combined with HSP90 inhibition.
Collapse
|
4
|
Huang Y, Wang Y, Tang J, Qin S, Shen X, He S, Ju S. CAM-DR: Mechanisms, Roles and Clinical Application in Tumors. Front Cell Dev Biol 2021; 9:698047. [PMID: 34295898 PMCID: PMC8290360 DOI: 10.3389/fcell.2021.698047] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Accepted: 06/08/2021] [Indexed: 12/14/2022] Open
Abstract
Despite the continuous improvement of various therapeutic techniques, the overall prognosis of tumors has been significantly improved, but malignant tumors in the middle and advanced stages still cannot be completely cured. It is now evident that cell adhesion-mediated resistance (CAM-DR) limits the success of cancer therapies and is a great obstacle to overcome in the clinic. The interactions between tumor cells and extracellular matrix (ECM) molecules or adjacent cells may play a significant role in initiating the intracellular signaling pathways that are associated with cell proliferation, survival upon binding to their ligands. Recent studies illustrate that these adhesion-related factors may contribute to the survival of cancer cells after chemotherapeutic therapy, advantageous to resistant cells to proliferate and develop multiple mechanisms of drug resistance. In this review, we focus on the molecular basis of these interactions and the main signal transduction pathways that are involved in the enhancement of the cancer cells’ survival. Furthermore, therapies targeting interactions between cancer cells and their environment to enhance drug response or prevent the emergence of drug resistance will also be discussed.
Collapse
Affiliation(s)
- Yuejiao Huang
- Medical School, Nantong University, Nantong, China.,Department of Medical Oncology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Yuchan Wang
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong, China
| | - Jie Tang
- Department of Pathogenic Biology, School of Medicine, Nantong University, Nantong, China
| | - Shiyi Qin
- Medical School, Nantong University, Nantong, China.,Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Xianjuan Shen
- Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| | - Song He
- Department of Pathology, Affiliated Tumor Hospital of Nantong University, Nantong, China
| | - Shaoqing Ju
- Medical School, Nantong University, Nantong, China.,Department of Laboratory Medicine, Affiliated Hospital of Nantong University, Nantong, China
| |
Collapse
|
5
|
Sueur G, Boutet A, Gotanègre M, Mansat-De Mas V, Besson A, Manenti S, Bertoli S. STAT5-dependent regulation of CDC25A by miR-16 controls proliferation and differentiation in FLT3-ITD acute myeloid leukemia. Sci Rep 2020; 10:1906. [PMID: 32024878 PMCID: PMC7002454 DOI: 10.1038/s41598-020-58651-x] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/24/2019] [Accepted: 01/14/2020] [Indexed: 01/10/2023] Open
Abstract
We recently identified the CDC25A phosphatase as a key actor in proliferation and differentiation in acute myeloid leukemia expressing the FLT3-ITD mutation. In this paper we demonstrate that CDC25A level is controlled by a complex STAT5/miR-16 transcription and translation pathway working downstream of this receptor. First, we established by CHIP analysis that STAT5 is directly involved in FLT3-ITD-dependent CDC25A gene transcription. In addition, we determined that miR-16 expression is repressed by FLT3-ITD activity, and that STAT5 participates in this repression. In accordance with these results, miR-16 expression was significantly reduced in a panel of AML primary samples carrying the FLT3-ITD mutation when compared with FLT3wt cells. The expression of a miR-16 mimic reduced CDC25A protein and mRNA levels, and RNA interference-mediated down modulation of miR-16 restored CDC25A expression in response to FLT3-ITD inhibition. Finally, decreasing miR-16 expression partially restored the proliferation of cells treated with the FLT3 inhibitor AC220, while the expression of miR-16 mimic stopped this proliferation and induced monocytic differentiation of AML cells. In summary, we identified a FLT3-ITD/STAT5/miR-16/CDC25A axis essential for AML cell proliferation and differentiation.
Collapse
Affiliation(s)
- Gabrielle Sueur
- Cancer Research Center of Toulouse (CRCT), INSERM U1037, CNRS ERL5294, University of Toulouse, Toulouse, France.,Equipe labellisée La Ligue contre le Cancer 2016, Toulouse, France
| | - Alison Boutet
- Cancer Research Center of Toulouse (CRCT), INSERM U1037, CNRS ERL5294, University of Toulouse, Toulouse, France.,Equipe labellisée La Ligue contre le Cancer 2016, Toulouse, France
| | - Mathilde Gotanègre
- Cancer Research Center of Toulouse (CRCT), INSERM U1037, CNRS ERL5294, University of Toulouse, Toulouse, France
| | - Véronique Mansat-De Mas
- Cancer Research Center of Toulouse (CRCT), INSERM U1037, CNRS ERL5294, University of Toulouse, Toulouse, France.,Equipe labellisée La Ligue contre le Cancer 2016, Toulouse, France.,Laboratoire d'hématologie, Institut Universitaire du Cancer de Toulouse-Oncopole, Toulouse, France.,Université Toulouse III Paul Sabatier, Toulouse, France
| | - Arnaud Besson
- Laboratoire de Biologie Cellulaire et Moléculaire du Contrôle de la Prolifération, Centre de Biologie Intégrative, Université de Toulouse, UPS and CNRS, Toulouse, France
| | - Stéphane Manenti
- Cancer Research Center of Toulouse (CRCT), INSERM U1037, CNRS ERL5294, University of Toulouse, Toulouse, France. .,Equipe labellisée La Ligue contre le Cancer 2016, Toulouse, France.
| | - Sarah Bertoli
- Cancer Research Center of Toulouse (CRCT), INSERM U1037, CNRS ERL5294, University of Toulouse, Toulouse, France. .,Equipe labellisée La Ligue contre le Cancer 2016, Toulouse, France. .,Université Toulouse III Paul Sabatier, Toulouse, France. .,Service d'hématologie, Centre Hospitalier Universitaire de Toulouse, Institut Universitaire du Cancer de Toulouse-Oncopole (IUCT-O), Toulouse, France.
| |
Collapse
|
6
|
Song B, Chen Y, Liu Y, Wan C, Zhang L, Zhang W. NPAS2 regulates proliferation of acute myeloid leukemia cells via CDC25A-mediated cell cycle progression and apoptosis. J Cell Biochem 2019; 120:8731-8741. [PMID: 30536616 DOI: 10.1002/jcb.28160] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/11/2018] [Accepted: 11/08/2018] [Indexed: 01/24/2023]
Abstract
Promoted proliferation and associated suppression of apoptosis at various stages of myeloid differentiation are well-known features of acute myeloid leukemia (AML), but understanding of the molecular processes involved remains limited. As a crucial circadian agent, neuronal PAS domain protein 2 (NPAS2) is widely recognized as a promising predictor of clinical outcome in various malignancies. Nevertheless, the understanding of its influence on AML is insufficient. Using KD cells and expression assays, we carried out detailed investigation of the role of NPAS2 in AML in vivo and in vitro. Firstly, we found that NPAS2 expression was elevated in AML cells both in vivo and in vitro. NPAS2 knockdown via lentiviral infection clearly suppressed proliferation of MV4-11 and MOLM-14 cells. Additionally, NPAS2 knockdown caused G1/S cell cycle arrest (CCA), which inhibited CDC25A expression. Moreover, NPAS2 knockdown promoted cell death, as evidenced by increased caspase-3 cleavage, and change in Bcl2/Bax production. Excessive CDC25A expression eliminated G1/S CCA triggered by NPAS2 knockdown and death of NPAS2 knocked down MOLM and MV4-11 cells. The expression of CDC25A was stabilized by NPAS2, which induced cell cycle progression and participated in suppression of cell death by modulating caspase-3 cleavage, and expression of Bcl2/Bax. We therefore indicated NPAS2 to be a crucial modulator of survival as well as proliferation. Our research sheds light on the etiology of the proliferation of promyelocytes modulated via NPAS2 with regard to AML.
Collapse
Affiliation(s)
- Bin Song
- Department of Hematology, Affiliated Taihe Hospital of Xi'an Jiaotong University Health Science Center, Shiyan, China
| | - Yan Chen
- Department of Hematology, Affiliated Taihe Hospital of Xi'an Jiaotong University Health Science Center, Shiyan, China
| | - Yuhong Liu
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Chucheng Wan
- Department of Hematology, Affiliated Taihe Hospital of Xi'an Jiaotong University Health Science Center, Shiyan, China
| | - Longjin Zhang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| | - Wanggang Zhang
- Department of Hematology, The Second Affiliated Hospital of Xi'an Jiaotong University, Xi'an, China
| |
Collapse
|
7
|
Johansen S, Brenner AK, Bartaula-Brevik S, Reikvam H, Bruserud Ø. The Possible Importance of β3 Integrins for Leukemogenesis and Chemoresistance in Acute Myeloid Leukemia. Int J Mol Sci 2018; 19:ijms19010251. [PMID: 29342970 PMCID: PMC5796198 DOI: 10.3390/ijms19010251] [Citation(s) in RCA: 36] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/2017] [Revised: 12/20/2017] [Accepted: 01/08/2018] [Indexed: 12/25/2022] Open
Abstract
Acute myeloid leukemia (AML) is an aggressive bone marrow malignancy where the immature leukemia cells communicate with neighboring cells through constitutive cytokine release and through their cell surface adhesion molecules. The primary AML cells express various integrins. These heterodimeric molecules containing an α and a β chain are cell surface molecules that bind extracellular matrix molecules, cell surface molecules and soluble mediators. The β3 integrin (ITGB3) chain can form heterodimers only with the two α chains αIIb and αV. These integrins are among the most promiscuous and bind to a large number of ligands, including extracellular matrix molecules, cell surface molecules and soluble mediators. Recent studies suggest that the two β3 integrins are important for leukemogenesis and chemosensitivity in human AML. Firstly, αIIb and β3 are both important for adhesion of AML cells to vitronectin and fibronectin. Secondly, β3 is important for the development of murine AML and also for the homing and maintenance of the proliferation for xenografted primary human AML cells, and for maintaining a stem cell transcriptional program. These last effects seem to be mediated through Syk kinase. The β3 expression seems to be regulated by HomeboxA9 (HoxA9) and HoxA10, and the increased β3 expression then activates spleen tyrosine kinase (Syk) and thereby contributes to cytokine hypersensitivity and activation of β2 integrins. Finally, high integrin αV/β3 expression is associated with an adverse prognosis in AML and decreased sensitivity to the kinase inhibitor sorafenib; this integrin can also be essential for osteopontin-induced sorafenib resistance in AML. In the present article, we review the experimental and clinical evidence for a role of β3 integrins for leukemogenesis and chemosensitivity in AML.
Collapse
MESH Headings
- Animals
- Cell Transformation, Neoplastic/genetics
- Cell Transformation, Neoplastic/metabolism
- Disease Models, Animal
- Drug Resistance, Neoplasm/genetics
- Gene Expression Regulation, Neoplastic
- Humans
- Integrin beta3/chemistry
- Integrin beta3/genetics
- Integrin beta3/metabolism
- Integrins/chemistry
- Integrins/genetics
- Integrins/metabolism
- Leukemia, Myeloid, Acute/drug therapy
- Leukemia, Myeloid, Acute/etiology
- Leukemia, Myeloid, Acute/metabolism
- Leukemia, Myeloid, Acute/pathology
- Ligands
- Multigene Family
- Prognosis
- Protein Binding
- Signal Transduction
Collapse
Affiliation(s)
- Silje Johansen
- Section for Hematology, Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway.
| | - Annette K Brenner
- Section for Hematology, Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway.
- Section for Hematology, Institute of Clinical Science, University of Bergen, 5007 Bergen, Norway.
| | - Sushma Bartaula-Brevik
- Section for Hematology, Institute of Clinical Science, University of Bergen, 5007 Bergen, Norway.
| | - Håkon Reikvam
- Section for Hematology, Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway.
- Section for Hematology, Institute of Clinical Science, University of Bergen, 5007 Bergen, Norway.
| | - Øystein Bruserud
- Section for Hematology, Department of Medicine, Haukeland University Hospital, N-5021 Bergen, Norway.
- Section for Hematology, Institute of Clinical Science, University of Bergen, 5007 Bergen, Norway.
| |
Collapse
|
8
|
Eldawud R, Wagner A, Dong C, Stueckle TA, Rojanasakul Y, Dinu CZ. Carbon nanotubes physicochemical properties influence the overall cellular behavior and fate. NANOIMPACT 2018; 9:72-84. [PMID: 31544167 PMCID: PMC6753956 DOI: 10.1016/j.impact.2017.10.006] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/29/2023]
Abstract
The unique properties of single walled carbon nanotubes (SWCNTs) make them viable candidates for versatile implementation in the next generation of biomedical devices for targeted delivery of chemotherapeutic agents or cellular-sensing probes. Such implementation requires user-tailored changes in SWCNT's physicochemical characteristics to allow for efficient cellular integration while maintaining nanotubes' functionality. However, isolated reports showed that user-tailoring could induce deleterious effects in exposed cells, from decrease in cellular proliferation, to changes in cellular adhesion, generation of reactive oxygen species or phenotypical variations, just to name a few. Before full implementation of SWCNTs is achieved, their toxicological profiles need to be mechanistically correlated with their physicochemical properties to determine how the induced cellular fate is related to the exposure conditions or samples' characteristics. Our study provides a comprehensive analysis of the synergistic cyto- and genotoxic effects resulted from short-term exposure of human lung epithelial cells to pristine (as manufactured) and user-tailored SWCNTs, as a function of their physicochemical properties. Specifically, through a systematic approach we are correlating the nanotube uptake and nanotube-induced cellular changes to the sample's physicochemical characteristics (e.g., metal impurities, length, agglomerate size, surface area, dispersion, and surface functionalization). By identifying changes in active hallmarks involved in cell-cell connections and maintaining epithelial layer integrity, we also determine the role that short-term exposure to SWCNTs plays in the overall cellular fate and cellular transformation. Lastly, we assess cellular structure-function relationships to identify non-apoptotic pathways induced by SWCNTs exposure that could however lead to changes in cellular behavior and cellular transformation. Our results show that the degree of cell transformation is a function of the physicochemical properties of the SWCNT, with the nanotube with higher length, higher metal content and larger agglomerate size reducing cell viability to a larger extent. Such changes in cell viability are also complemented by changes in cell structure, cycle and cell-cell interactions, all responsible for maintaining cell fate.
Collapse
Affiliation(s)
- Reem Eldawud
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA
| | - Alixandra Wagner
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA
| | - Chenbo Dong
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA
| | - Todd A. Stueckle
- Health Effects Laboratory Division, National Institute for Occupational Safety and Health, Morgantown, WV 26505, USA
| | - Yon Rojanasakul
- Department of Pharmaceutical Sciences, West Virginia University, WV 26506, USA
| | - Cerasela Zoica Dinu
- Department of Chemical and Biomedical Engineering, West Virginia University, Morgantown, WV 26506, USA
- Department of Pharmaceutical Sciences, West Virginia University, WV 26506, USA
| |
Collapse
|
9
|
Chen D, Long M, Xiao B, Xiong Y, Chen H, Chen Y, Kuang Z, Li M, Wu Y, Rock DL, Gong D, Wang Y, He H, Liu F, Luo S, Hao W. Transcriptomic profiles of human foreskin fibroblast cells in response to orf virus. Oncotarget 2017; 8:58668-58685. [PMID: 28938587 PMCID: PMC5601683 DOI: 10.18632/oncotarget.17417] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/18/2017] [Accepted: 03/20/2017] [Indexed: 11/25/2022] Open
Abstract
Orf virus has been utilized as a safe and efficient viral vector against not only diverse infectious diseases, but also against tumors. However, the nature of the genes triggered by the vector in human cells is poorly characterized. Using RNA sequencing technology, we compared specific changes in the transcriptomic profiles in human foreskin fibroblast cells following infection by the orf virus. The results indicated that orf virus upregulates or downregulates expression of a variety of genes, including genes involved in antiviral immune response, apoptosis, cell cycle and a series of signaling pathways, such as the IFN and p53-signaling pathways. The orf virus stimulates or inhibits immune gene expression such as chemokines, chemokine receptors, cytokines, cytokine receptors, and molecules involved in antigen uptake and processing after infection. Expression of pro-apoptotic genes increased at 8 hours post-infection. The p53 signaling pathway was activated to induce apoptosis at the same time. However, the cell cycle program was promoted after infection, which may be due to the immunomodulatory genes of the orf virus. This presents the first description of transcription profile changes in human foreskin fibroblast cells after orf virus infection and provides an in-depth analysis of the interaction between the host and orf virus. These data offer new insights into the understanding of the mechanisms of infection by orf virus and identify potential targets for future studies.
Collapse
Affiliation(s)
- Daxiang Chen
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China
| | - Mingjian Long
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China
| | - Bin Xiao
- Department of Laboratory Medicine, Guangzhou General Hospital of Guangzhou Military Command of PLA, Guangzhou, 510010, P.R. China
| | - Yufeng Xiong
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China
| | - Huiqin Chen
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China
| | - Yu Chen
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China
| | - Zhenzhan Kuang
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China
| | - Ming Li
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China.,Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, P.R. China
| | - Yingsong Wu
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China.,Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, P.R. China
| | - Daniel L Rock
- Department of Pathobiology, College of Veterinary Medicine, University of Illinois at Champaign-Urbana, Urbana, IL 61802 USA
| | - Daoyuan Gong
- Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Chancheng District, Foshan, Guangdong Province, 528000 P.R. China
| | - Yong Wang
- Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Chancheng District, Foshan, Guangdong Province, 528000 P.R. China
| | - Haijian He
- Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Chancheng District, Foshan, Guangdong Province, 528000 P.R. China
| | - Fang Liu
- Department of Pathophysiology, School of Stomatology and Medicine, Foshan University, Chancheng District, Foshan, Guangdong Province, 528000 P.R. China
| | - Shuhong Luo
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China.,Department of Laboratory Medicine, School of Stomatology and Medicine, Foshan University, Chancheng District, Foshan, Guangdong Province, 528000 P.R. China
| | - Wenbo Hao
- Institute of Antibody Engineering, School of Laboratory Medicine and Biotechnology, Southern Medical University, Guangzhou, 510515, P.R. China.,Guangdong Provincial Key Laboratory of Tropical Disease Research, School of Public Health, Southern Medical University, Guangzhou, 510515, P.R. China
| |
Collapse
|
10
|
CDC25 Inhibition in Acute Myeloid Leukemia-A Study of Patient Heterogeneity and the Effects of Different Inhibitors. Molecules 2017; 22:molecules22030446. [PMID: 28287460 PMCID: PMC6155411 DOI: 10.3390/molecules22030446] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2017] [Revised: 03/01/2017] [Accepted: 03/06/2017] [Indexed: 12/15/2022] Open
Abstract
Cell division cycle 25 (CDC25) protein phosphatases regulate cell cycle progression through the activation of cyclin-dependent kinases (CDKs), but they are also involved in chromatin modulation and transcriptional regulation. CDC25 inhibition is regarded as a possible therapeutic strategy for the treatment of human malignancies, including acute myeloid leukemia (AML). We investigated the in vitro effects of CDC25 inhibitors on primary human AML cells derived from 79 unselected patients in suspension cultures. Both the previously well-characterized CDC25 inhibitor NSC95397, as well as five other inhibitors (BN82002 and the novel small molecular compounds ALX1, ALX2, ALX3, and ALX4), only exhibited antiproliferative effects for a subset of patients when tested alone. These antiproliferative effects showed associations with differences in genetic abnormalities and/or AML cell differentiation. However, the responders to CDC25 inhibition could be identified by analysis of global gene expression profiles. The differentially expressed genes were associated with the cytoskeleton, microtubules, and cell signaling. The constitutive release of 28 soluble mediators showed a wide variation among patients and this variation was maintained in the presence of CDC25 inhibition. Finally, NSC95397 had no or only minimal effects on AML cell viability. In conclusion, CDC25 inhibition has antiproliferative effects on primary human AML cells for a subset of patients, and these patients can be identified by gene expression profiling.
Collapse
|
11
|
Dozier C, Mazzolini L, Cénac C, Froment C, Burlet-Schiltz O, Besson A, Manenti S. CyclinD-CDK4/6 complexes phosphorylate CDC25A and regulate its stability. Oncogene 2017; 36:3781-3788. [PMID: 28192398 DOI: 10.1038/onc.2016.506] [Citation(s) in RCA: 37] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2016] [Revised: 10/28/2016] [Accepted: 12/13/2016] [Indexed: 12/27/2022]
Abstract
The phosphatase CDC25A is a key regulator of cell cycle progression by dephosphorylating and activating cyclin-CDK complexes. CDC25A is an unstable protein expressed from G1 until mitosis. CDC25A overexpression, which can be caused by stabilization of the protein, accelerates the G1/S and G2/M transitions, leading to genomic instability and promoting tumorigenesis. Thus, controlling CDC25A protein levels by regulating its stability is a critical mechanism for timing cell cycle progression and to maintain genomic integrity. Herein, we show that CDC25A is phosphorylated on Ser40 throughout the cell cycle and that this phosphorylation is established during the progression from G1 to S phase. We demonstrate that CyclinD-CDK4/CDK6 complexes mediate the phosphorylation of CDC25A on Ser40 during G1 and that these complexes directly phosphorylate this residue in vitro. Importantly, we also find that CyclinD1-CDK4 decreases CDC25A stability in a ßTrCP-dependent manner and that Ser40 and Ser88 phosphorylations contribute to this regulation. Thus our results identify cyclinD-CDK4/6 complexes as novel regulators of CDC25A stability during G1 phase, generating a negative feedback loop allowing control of the G1/S transition.
Collapse
Affiliation(s)
- C Dozier
- Cancer Research Center of Toulouse, INSERM UMR1037/Université Toulouse III Paul Sabatier, Toulouse, France.,Equipe labellisée Ligue Contre le Cancer, CNRS ERL5294, Toulouse, France
| | - L Mazzolini
- Cancer Research Center of Toulouse, INSERM UMR1037/Université Toulouse III Paul Sabatier, Toulouse, France.,Equipe labellisée Ligue Contre le Cancer, CNRS ERL5294, Toulouse, France
| | - C Cénac
- Cancer Research Center of Toulouse, INSERM UMR1037/Université Toulouse III Paul Sabatier, Toulouse, France
| | - C Froment
- Institut de Pharmacologie et de Biologie Structurale, CNRS UMR5089, Université Toulouse, Toulouse, France
| | - O Burlet-Schiltz
- Institut de Pharmacologie et de Biologie Structurale, CNRS UMR5089, Université Toulouse, Toulouse, France
| | - A Besson
- Cancer Research Center of Toulouse, INSERM UMR1037/Université Toulouse III Paul Sabatier, Toulouse, France
| | - S Manenti
- Cancer Research Center of Toulouse, INSERM UMR1037/Université Toulouse III Paul Sabatier, Toulouse, France.,Equipe labellisée Ligue Contre le Cancer, CNRS ERL5294, Toulouse, France
| |
Collapse
|
12
|
Ouyang Y, Zhong F, Wang Q, Ding L, Zhang P, Chen L, Wang Y, Cheng C. DIXDC1 promotes tumor proliferation and cell adhesion mediated drug resistance (CAM-DR) via enhancing p-Akt in Non-Hodgkin's lymphomas. Leuk Res 2016; 50:104-111. [PMID: 27701018 DOI: 10.1016/j.leukres.2016.09.011] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/06/2016] [Revised: 09/04/2016] [Accepted: 09/07/2016] [Indexed: 12/20/2022]
Abstract
DIX domain containing 1 (DIXDC1), is a human homolog of Ccd1, a DIX domain containing protein in zebrafish. The present study was undertaken to determine the expression and biologic function of DIXDC1 in Non-Hodgkin's lymphoma (NHL). Clinically, we detected that the expression of DIXDC1 was significantly lower in the indolent lymphomas compared with the progressive lymphomas by immunohistochemistry analysis. Functionally, we found that DIXDC1 could promote cell proliferation via modulating cell cycle progression and PI3K/AKT signaling pathway in NHLs. Moreover, we confirmed that DIXDC1 was involved in the process of lymphoma cell adhesion mediated drug resistance (CAM-DR). Adhesion to fibronectin (FN) or HS-5 up-regulated DIXDC1 expression, and up-regulation of DIXDC1 resulted in an increased expression of p-AKT, which promoted CAM-DR. Our finding supports the role of DIXDC1 in cell proliferation, cell cycle and CAM-DR in NHLs. We propose that inhibition of DIXDC1 expression may be a novel therapeutic approach for NHLs patients, and it may be a target for drug resistance.
Collapse
Affiliation(s)
- Yu Ouyang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, Jiangsu, China
| | - Fei Zhong
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, Jiangsu, China
| | - Qiru Wang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, Jiangsu, China
| | - Linlin Ding
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, Jiangsu, China
| | - Peipei Zhang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, Jiangsu, China
| | - Lingling Chen
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, Jiangsu, China
| | - Yuchan Wang
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, Jiangsu, China.
| | - Chun Cheng
- Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong 226001, Jiangsu, China.
| |
Collapse
|
13
|
Huang Y, Xu X, Ji L, Wang Y, Wang S, Tang J, Huang X, Yang X, He Y, He S, Cheng C. Expression of far upstream element binding protein 1 in B‑cell non‑Hodgkin lymphoma is correlated with tumor growth and cell‑adhesion mediated drug resistance. Mol Med Rep 2016; 14:3759-68. [PMID: 27599538 DOI: 10.3892/mmr.2016.5718] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/08/2015] [Accepted: 08/09/2016] [Indexed: 11/05/2022] Open
Abstract
Cell adhesion‑mediated drug resistance (CAM‑DR) remains a major obstacle to the effectiveness of chemotherapeutic treatment of lymphoma. Far upstream element binding protein 1 (FBP1) is a multifunctional protein that is highly expressed in proliferating cells of several solid neoplasms; however, its expression and biological function in B‑cell lymphoma is largely unknown. FBP1 expression in both reactive lymphoid tissues and several B‑cell lymphomas, including follicular lymphoma and diffuse large B‑cell lymphoma were detected by immunohistochemistry analysis. FBP1 expression in B‑cell lymphoma was also associated with poor survival outcomes. Functionally, small interfering RNA‑mediated silencing of FBP1 was able to inhibit the proliferation of B‑cell lymphoma cells, resulting in G0/G1 phase cell cycle arrest. Furthermore, results of a cell adhesion assay demonstrated that adhesion to fibronectin or bone marrow stromal cells induced FBP1 expression, which in turn facilitated cell adhesion. Finally, FBP1 knockdown reversed CAM‑DR. These findings support a role for FBP1 in non‑Hodgkin lymphoma cell proliferation, adhesion and drug resistance, and may lead to the generation of a novel therapeutic approach targeting this molecule.
Collapse
Affiliation(s)
- Yuejiao Huang
- Department of Oncology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu 226361, P.R. China
| | - Xiaohong Xu
- Department of Oncology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu 226361, P.R. China
| | - Lili Ji
- Department of Pathology, Medical College, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yuchan Wang
- Department of Oncology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu 226361, P.R. China
| | - Shitao Wang
- Department of Pathology, Medical College, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Jie Tang
- Department of Immunology, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xianting Huang
- Department of Immunology, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Xiaojing Yang
- Department of Immunology, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Yunhua He
- Department of Immunology, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu 226001, P.R. China
| | - Song He
- Department of Pathology, Affiliated Cancer Hospital of Nantong University, Nantong, Jiangsu 226361, P.R. China
| | - Chun Cheng
- Department of Immunology, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target, Medical College, Nantong University, Nantong, Jiangsu 226001, P.R. China
| |
Collapse
|
14
|
Bertoli S, Boutzen H, David L, Larrue C, Vergez F, Fernandez-Vidal A, Yuan L, Hospital MA, Tamburini J, Demur C, Delabesse E, Saland E, Sarry JE, Galcera MO, Mansat-De Mas V, Didier C, Dozier C, Récher C, Manenti S. CDC25A governs proliferation and differentiation of FLT3-ITD acute myeloid leukemia. Oncotarget 2016; 6:38061-78. [PMID: 26515730 PMCID: PMC4741984 DOI: 10.18632/oncotarget.5706] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2015] [Accepted: 10/06/2015] [Indexed: 11/25/2022] Open
Abstract
We investigated cell cycle regulation in acute myeloid leukemia cells expressing the FLT3-ITD mutated tyrosine kinase receptor, an underexplored field in this disease. Upon FLT3 inhibition, CDC25A mRNA and protein were rapidly down-regulated, while levels of other cell cycle proteins remained unchanged. This regulation was dependent on STAT5, arguing for FLT3-ITD-dependent transcriptional regulation of CDC25A. CDC25 inhibitors triggered proliferation arrest and cell death of FLT3-ITD as well as FLT3-ITD/TKD AC-220 resistant cells, but not of FLT3-wt cells. Consistently, RNA interference-mediated knock-down of CDC25A reduced the proliferation of FLT3-ITD cell lines. Finally, the clonogenic capacity of primary FLT3-ITD AML cells was reduced by the CDC25 inhibitor IRC-083864, while FLT3-wt AML and normal CD34+ myeloid cells were unaffected. In good agreement, in a cohort of 100 samples from AML patients with intermediate-risk cytogenetics, high levels of CDC25A mRNA were predictive of higher clonogenic potential in FLT3-ITD+ samples, not in FLT3-wt ones.Importantly, pharmacological inhibition as well as RNA interference-mediated knock-down of CDC25A also induced monocytic differentiation of FLT3-ITD positive cells, as judged by cell surface markers expression, morphological modifications, and C/EBPα phosphorylation. CDC25 inhibition also re-induced monocytic differentiation in primary AML blasts carrying the FLT3-ITD mutation, but not in blasts expressing wild type FLT3. Altogether, these data identify CDC25A as an early cell cycle transducer of FLT3-ITD oncogenic signaling, and as a promising target to inhibit proliferation and re-induce differentiation of FLT3-ITD AML cells.
Collapse
Affiliation(s)
- Sarah Bertoli
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France.,Hematology Department, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France
| | - Helena Boutzen
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| | - Laure David
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| | - Clément Larrue
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France.,Institut Cochin, Université Paris Descartes, CNRS UMR 8104, INSERM U 1016, Paris, France
| | - François Vergez
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France.,Hematology Laboratory, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France
| | - Anne Fernandez-Vidal
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| | - Lingli Yuan
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| | - Marie-Anne Hospital
- Institut Cochin, Université Paris Descartes, CNRS UMR 8104, INSERM U 1016, Paris, France
| | - Jérôme Tamburini
- Institut Cochin, Université Paris Descartes, CNRS UMR 8104, INSERM U 1016, Paris, France
| | - Cécile Demur
- Hematology Laboratory, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France
| | - Eric Delabesse
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France.,Hematology Laboratory, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France
| | - Estelle Saland
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| | - Jean-Emmanuel Sarry
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| | | | - Véronique Mansat-De Mas
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France.,Hematology Laboratory, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France
| | - Christine Didier
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| | - Christine Dozier
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| | - Christian Récher
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France.,Hematology Department, Institut Universitaire du Cancer Toulouse - Oncopole, Toulouse, France
| | - Stéphane Manenti
- Cancer Research Center of Toulouse, Inserm UMR 1037, CNRS ERL 5294, Université de Toulouse, Oncopole, Toulouse, France
| |
Collapse
|
15
|
He S, Huang Y, Wang Y, Tang J, Song Y, Yu X, Ma J, Wang S, Yin H, Li Q, Ji L, Xu X. Histamine-releasing factor/translationally controlled tumor protein plays a role in induced cell adhesion, apoptosis resistance and chemoresistance in non-Hodgkin lymphomas. Leuk Lymphoma 2015; 56:2153-61. [PMID: 25363345 DOI: 10.3109/10428194.2014.981173] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Mounting evidence has proved that cellular adhesion confers resistance to chemotherapy in multiple lymphomas. The molecular mechanism underlying cell adhesion-mediated drug resistance (CAM-DR) is, however, poorly understood. In this study, we investigated the expression and biologic function of histamine-releasing factor (HRF) in non-Hodgkin lymphomas (NHLs). Clinically, by immunohistochemistry analysis we observed obvious up-regulation of HRF in NHLs including diffuse large B-cell lymphoma (DLBCL), follicular lymphoma (FL) and natural killer (NK)/T-cell lymphoma. Functionally, overexpression and knockdown of HRF demonstrated the antiapoptotic effect of HRF in NHL cells, which may be associated with activation of the p-CREB/BCL-2 signaling pathway. Moreover, cell adhesion assay demonstrated that adhesion to fibronectin (FN) or HS-5 up-regulated HRF expression, while knockdown of HRF resulted in decreased cell adhesion, which led to reversed CAM-DR. Our finding supports the role of HRF in NHL cell apoptosis, adhesion and drug resistance, and may provide a clinical therapeutic target for CAM-DR in NHL.
Collapse
Affiliation(s)
- Song He
- Department of Oncology, Affiliated Cancer Hospital of Nantong University, Jiangsu Province Key Laboratory for Inflammation and Molecular Drug Target , Nantong, Jiangsu , China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
16
|
Progress in RNAi-mediated Molecular Therapy of Acute and Chronic Myeloid Leukemia. MOLECULAR THERAPY. NUCLEIC ACIDS 2015; 4:e240. [DOI: 10.1038/mtna.2015.13] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/13/2015] [Accepted: 03/26/2015] [Indexed: 02/08/2023]
|
17
|
Brenner AK, Reikvam H, Lavecchia A, Bruserud Ø. Therapeutic targeting the cell division cycle 25 (CDC25) phosphatases in human acute myeloid leukemia--the possibility to target several kinases through inhibition of the various CDC25 isoforms. Molecules 2014; 19:18414-47. [PMID: 25397735 PMCID: PMC6270710 DOI: 10.3390/molecules191118414] [Citation(s) in RCA: 63] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2014] [Revised: 10/28/2014] [Accepted: 11/02/2014] [Indexed: 01/26/2023] Open
Abstract
The cell division cycle 25 (CDC25) phosphatases include CDC25A, CDC25B and CDC25C. These three molecules are important regulators of several steps in the cell cycle, including the activation of various cyclin-dependent kinases (CDKs). CDC25s seem to have a role in the development of several human malignancies, including acute myeloid leukemia (AML); and CDC25 inhibition is therefore considered as a possible anticancer strategy. Firstly, upregulation of CDC25A can enhance cell proliferation and the expression seems to be controlled through PI3K-Akt-mTOR signaling, a pathway possibly mediating chemoresistance in human AML. Loss of CDC25A is also important for the cell cycle arrest caused by differentiation induction of malignant hematopoietic cells. Secondly, high CDC25B expression is associated with resistance against the antiproliferative effect of PI3K-Akt-mTOR inhibitors in primary human AML cells, and inhibition of this isoform seems to reduce AML cell line proliferation through effects on NFκB and p300. Finally, CDC25C seems important for the phenotype of AML cells at least for a subset of patients. Many of the identified CDC25 inhibitors show cross-reactivity among the three CDC25 isoforms. Thus, by using such cross-reactive inhibitors it may become possible to inhibit several molecular events in the regulation of cell cycle progression and even cytoplasmic signaling, including activation of several CDKs, through the use of a single drug. Such combined strategies will probably be an advantage in human cancer treatment.
Collapse
Affiliation(s)
- Annette K Brenner
- Section for Hematology, Institute of Clinical Science, Faculty of Medicine and Dentistry, University of Bergen, Bergen, 5021, Norway
| | - Håkon Reikvam
- Section for Hematology, Institute of Clinical Science, Faculty of Medicine and Dentistry, University of Bergen, Bergen, 5021, Norway
| | - Antonio Lavecchia
- "Drug Discovery" Laboratory, Department of Pharmacy, University of Naples Federico II, Naples 80131, Italy
| | - Øystein Bruserud
- Section for Hematology, Institute of Clinical Science, Faculty of Medicine and Dentistry, University of Bergen, Bergen, 5021, Norway.
| |
Collapse
|
18
|
Wang Y, Huang Y, Xu X, Tang J, Huang X, Zhu J, Liu J, Miao X, Wu Y, Yang F, Ji L, He S. Expression of small glutamine-rich TPR-containing protein A (SGTA) in Non-Hodgkin's Lymphomas promotes tumor proliferation and reverses cell adhesion-mediated drug resistance (CAM-DR). Leuk Res 2014; 38:955-63. [PMID: 24974147 DOI: 10.1016/j.leukres.2014.05.013] [Citation(s) in RCA: 14] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2013] [Revised: 05/09/2014] [Accepted: 05/21/2014] [Indexed: 12/16/2022]
Abstract
The expression and biologic function of SGTA in Non-Hodgkin's Lymphomas (NHL) was investigated in this study. Clinically, by immunohistochemistry analysis we detected SGTA expression in both reactive lymphoid tissues and NHL tissues. In addition, we also correlated high expression of SGTA with poor prognosis. Functionally, SGTA expression was positively related with cell proliferation and negative related with cell adhesion. Finally, SGTA knockdown induced adhesion-mediated drug resistance. Our finding supports a role of SGTA in NHL cell proliferation, adhesion and drug resistance, and it may pave the way for a novel therapeutic approach for CAM-DR in NHL.
Collapse
Affiliation(s)
- Yuchan Wang
- Department of Pathology, Affiliated Cancer Hospital of Nantong University, Nantong 226361, Jiangsu, China
| | - Yuejiao Huang
- Department of Pathology, Medical College, Nantong University, Nantong 226001, Jiangsu, China
| | - Xiaohong Xu
- Department of Pathology, Affiliated Cancer Hospital of Nantong University, Nantong 226361, Jiangsu, China
| | - Jie Tang
- Department of Pathology, Medical College, Nantong University, Nantong 226001, Jiangsu, China
| | - Xianting Huang
- Department of Pathology, Medical College, Nantong University, Nantong 226001, Jiangsu, China
| | - Junya Zhu
- Department of Pathology, Medical College, Nantong University, Nantong 226001, Jiangsu, China
| | - Jing Liu
- Department of Pathology, Medical College, Nantong University, Nantong 226001, Jiangsu, China
| | - Xiaobing Miao
- Department of Pathology, Affiliated Cancer Hospital of Nantong University, Nantong 226361, Jiangsu, China
| | - Yaxun Wu
- Department of Pathology, Affiliated Cancer Hospital of Nantong University, Nantong 226361, Jiangsu, China
| | - Fan Yang
- Department of Pediatrics, Medical College, Nantong University, Nantong 226001, Jiangsu, China
| | - Lili Ji
- Department of Pathology, Medical College, Nantong University, Nantong 226001, Jiangsu, China.
| | - Song He
- Department of Pathology, Affiliated Cancer Hospital of Nantong University, Nantong 226361, Jiangsu, China.
| |
Collapse
|
19
|
Lavecchia A, Di Giovanni C, Pesapane A, Montuori N, Ragno P, Martucci NM, Masullo M, De Vendittis E, Novellino E. Discovery of new inhibitors of Cdc25B dual specificity phosphatases by structure-based virtual screening. J Med Chem 2012; 55:4142-58. [PMID: 22524450 DOI: 10.1021/jm201624h] [Citation(s) in RCA: 39] [Impact Index Per Article: 3.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022]
Abstract
Cell division cycle 25 (Cdc25) proteins are highly conserved dual specificity phosphatases that regulate cyclin-dependent kinases and represent attractive drug targets for anticancer therapies. To discover more potent and diverse inhibitors of Cdc25 biological activity, virtual screening was performed by docking 2.1 million compounds into the Cdc25B active site. An initial subset of top-ranked compounds was selected and assayed, and 15 were found to have enzyme inhibition activity at micromolar concentration. Among these, four structurally diverse inhibitors with a different inhibition profile were found to inhibit human MCF-7, PC-3, and K562 cancer cell proliferation and significantly affect the cell cycle progression. A subsequent hierarchical similarity search with the most active reversible Cdc25B inhibitor found led to the identification of an additional set of 19 ligands, three of which were confirmed as Cdc25B inhibitors with IC(50) values of 7.9, 4.2, and 9.9 μM, respectively.
Collapse
Affiliation(s)
- Antonio Lavecchia
- Dipartimento di Chimica Farmaceutica e Tossicologica, Drug Discovery Laboratory, Università di Napoli Federico II, Via Domenico Montesano 49, 80131 Napoli, Italy.
| | | | | | | | | | | | | | | | | |
Collapse
|
20
|
Abstract
The JAK2(V617F) mutation is present in the majority of patients with polycythemia vera and one-half of those with essential thrombocythemia and primary myelofibrosis. JAK2(V617F) is a gain-of-function mutation resulting in constitutive JAK2 signaling involved in the pathogenesis of these diseases. JAK2(V617F) has been shown to promote S-phase entry. Here, we demonstrate that the CDC25A phosphatase, a key regulator of the G1/S cell-cycle transition, is constitutively overexpressed in JAK2(V617F)-positive cell lines, JAK2-mutated patient CD36(+) progenitors, and in vitro-differentiated proerythroblasts. Accordingly, CDC25A is overexpressed in BM and spleen of Jak2(V617F) knock-in mice compared with wild-type littermates. By using murine FDC-P1-EPOR and human HEL and SET-2 cell lines, we found that JAK2(V617F)-induced CDC25A up-regulation was caused neither by increased CDC25A transcription or stability nor by the involvement of its upstream regulators Akt and MAPK. Instead, our results suggest that CDC25A is regulated at the translational level through STAT5 and the translational initiation factor eIF2α. CDC25A inhibition reduces the clonogenic and proliferative potential of JAK2(V617F)-expressing cell lines and erythroid progenitors while moderately affecting normal erythroid differentiation. These results suggest that CDC25A deregulation may be involved in hematopoietic cells expansion in JAK2(V617F) patients, making this protein an attracting potential therapeutic target.
Collapse
|
21
|
Lamsoul I, Burande CF, Razinia Z, Houles TC, Menoret D, Baldassarre M, Erard M, Moog-Lutz C, Calderwood DA, Lutz PG. Functional and structural insights into ASB2alpha, a novel regulator of integrin-dependent adhesion of hematopoietic cells. J Biol Chem 2011; 286:30571-30581. [PMID: 21737450 DOI: 10.1074/jbc.m111.220921] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023] Open
Abstract
By providing contacts between hematopoietic cells and the bone marrow microenvironment, integrins are implicated in cell adhesion and thereby in control of cell fate of normal and leukemia cells. The ASB2 gene, initially identified as a retinoic acid responsive gene and a target of the promyelocytic leukemia retinoic acid receptor α oncoprotein in acute promyelocytic leukemia cells, encodes two isoforms, a hematopoietic-type (ASB2α) and a muscle-type (ASB2β) that are involved in hematopoietic and myogenic differentiation, respectively. ASB2α is the specificity subunit of an E3 ubiquitin ligase complex that targets filamins to proteasomal degradation. To examine the relationship of the ASB2α structure to E3 ubiquitin ligase function, functional assays and molecular modeling were performed. We show that ASB2α, through filamin A degradation, enhances adhesion of hematopoietic cells to fibronectin, the main ligand of β1 integrins. Furthermore, we demonstrate that a short N-terminal region specific to ASB2α, together with ankyrin repeats 1 to 10, is necessary for association of ASB2α with filamin A. Importantly, the ASB2α N-terminal region comprises a 9-residue segment with predicted structural homology to the filamin-binding motifs of migfilin and β integrins. Together, these data provide new insights into the molecular mechanisms of ASB2α binding to filamin.
Collapse
Affiliation(s)
- Isabelle Lamsoul
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 205 route de Narbonne, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - Clara F Burande
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 205 route de Narbonne, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - Ziba Razinia
- Department of Pharmacology and Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Thibault C Houles
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 205 route de Narbonne, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - Delphine Menoret
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 205 route de Narbonne, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - Massimiliano Baldassarre
- Department of Pharmacology and Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Monique Erard
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 205 route de Narbonne, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - Christel Moog-Lutz
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 205 route de Narbonne, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France
| | - David A Calderwood
- Department of Pharmacology and Interdepartmental Program in Vascular Biology and Transplantation, Yale University School of Medicine, New Haven, Connecticut 06520
| | - Pierre G Lutz
- Centre National de la Recherche Scientifique, Institut de Pharmacologie et de Biologie Structurale, 205 route de Narbonne, 31077 Toulouse, France; Université de Toulouse, Université Paul Sabatier, Institut de Pharmacologie et de Biologie Structurale, 31077 Toulouse, France.
| |
Collapse
|
22
|
Vispé S, DeVries L, Créancier L, Besse J, Bréand S, Hobson DJ, Svejstrup JQ, Annereau JP, Cussac D, Dumontet C, Guilbaud N, Barret JM, Bailly C. Triptolide is an inhibitor of RNA polymerase I and II-dependent transcription leading predominantly to down-regulation of short-lived mRNA. Mol Cancer Ther 2009; 8:2780-90. [PMID: 19808979 DOI: 10.1158/1535-7163.mct-09-0549] [Citation(s) in RCA: 135] [Impact Index Per Article: 8.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Triptolide, a natural product extracted from the Chinese plant Tripterygium wilfordii, possesses antitumor properties. Despite numerous reports showing the proapoptotic capacity and the inhibition of NF-kappaB-mediated transcription by triptolide, the identity of its cellular target is still unknown. To clarify its mechanism of action, we further investigated the effect of triptolide on RNA synthesis in the human non-small cell lung cancer cell line A549. Triptolide inhibited both total RNA and mRNA de novo synthesis, with the primary action being on the latter pool. We used 44K human pan-genomic DNA microarrays and identified the genes primarily affected by a short treatment with triptolide. Among the modulated genes, up to 98% are down-regulated, encompassing a large array of oncogenes including transcription factors and cell cycle regulators. We next observed that triptolide induced a rapid depletion of RPB1, the RNA polymerase II main subunit that is considered a hallmark of a transcription elongation blockage. However, we also show that triptolide does not directly interact with the RNA polymerase II complex nor does it damage DNA. We thus conclude that triptolide is an original pharmacologic inhibitor of RNA polymerase activity, affecting indirectly the transcription machinery, leading to a rapid depletion of short-lived mRNA, including transcription factors, cell cycle regulators such as CDC25A, and the oncogenes MYC and Src. Overall, the data shed light on the effect of triptolide on transcription, along with its novel potential applications in cancers, including acute myeloid leukemia, which is in part driven by the aforementioned oncogenic factors.
Collapse
Affiliation(s)
- Stéphane Vispé
- Centre de Recherche en Oncologie Expérimentale, Institut de Recherche Pierre Fabre, 3 rue des satellites, BP94244, Toulouse Cedex 4, 31432 France.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
23
|
Renner AG, Dos Santos C, Recher C, Bailly C, Créancier L, Kruczynski A, Payrastre B, Manenti S. Polo-like kinase 1 is overexpressed in acute myeloid leukemia and its inhibition preferentially targets the proliferation of leukemic cells. Blood 2009; 114:659-62. [PMID: 19458358 DOI: 10.1182/blood-2008-12-195867] [Citation(s) in RCA: 107] [Impact Index Per Article: 6.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Polo-like kinase 1 (Plk1) is a major mitotic regulator overexpressed in many solid tumors. Its role in hematopoietic malignancies is still poorly characterized. In this study, we demonstrate that Plk1 is highly expressed in leukemic cell lines, and overexpressed in a majority of samples from patients with acute myeloid leukemia compared with normal progenitors. A pharmacologic inhibitor, BI2536, blocks proliferation in established cell lines, and dramatically inhibits the clonogenic potential of leukemic cells from patients. Plk1 knockdown by small interfering RNA also blocked proliferation of leukemic cell lines and the clonogenic potential of primary cells from patients. Interestingly, normal primary hematopoietic progenitors are less sensitive to Plk1 inhibition than leukemic cells, whose proliferation is dramatically decreased by the inhibitor. These results highlight Plk1 as a potentially interesting therapeutic target for the treatment of acute myeloid leukemia.
Collapse
Affiliation(s)
- Annelies G Renner
- INSERM Unité 563-IFR30, Centre de Physiopathologie Toulouse-Purpan, Département Oncogenèse et Signalisation dans les cellules hématopoïétiques, Centre Hospitalier Universitaire (CHU) Purpan, Toulouse
| | | | | | | | | | | | | | | |
Collapse
|
24
|
|
25
|
A caspase-dependent cleavage of CDC25A generates an active fragment activating cyclin-dependent kinase 2 during apoptosis. Cell Death Differ 2008; 16:208-18. [PMID: 18927589 DOI: 10.1038/cdd.2008.142] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/06/2023] Open
Abstract
The cellular level of the CDC25A phosphatase is tightly regulated during both the normal and genotoxic-perturbed cell cycle. Here, we describe a caspase-dependent cleavage of this protein at residue D223 in non-genotoxic apoptotic conditions. This specific proteolysis generates a catalytically active C-terminal fragment that localizes to the nuclear compartment. Accumulation of this active CDC25A fragment leads to reduced inhibitory phosphorylation of the CDC25A substrate cyclin-dependent kinase 2 (CDK2) on Tyr15. Moreover, CDK2 was found stably associated with this fragment, as well as with an ectopically expressed CDC25A224-525 truncation mutant that mimicks the cleavage product. Ectopic expression of this mutant induced CDK2 Tyr15 dephosphorylation, whereas its catalytically inactive version did not. Finally, this 224-525 mutant initiated apoptosis when transfected into HeLa cells, whereas its catalytic inactive form did not. Altogether, this study demonstrates for the first time that caspase-dependent cleavage of CDC25A is a central step linking CDK2 activation with non-genotoxic apoptotic induction.
Collapse
|
26
|
Rodrigues MS, Reddy MM, Sattler M. Cell cycle regulation by oncogenic tyrosine kinases in myeloid neoplasias: from molecular redox mechanisms to health implications. Antioxid Redox Signal 2008; 10:1813-48. [PMID: 18593226 DOI: 10.1089/ars.2008.2071] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/23/2022]
Abstract
Neoplastic expansion of myeloid cells is associated with specific genetic changes that lead to chronic activation of signaling pathways, as well as altered metabolism. It has become increasingly evident that transformation relies on the interdependency of both events. Among the various genetic changes, the oncogenic BCR-ABL tyrosine kinase in patients with Philadelphia chromosome positive chronic myeloid leukemia (CML) has been a focus of extensive research. Transformation by this oncogene is associated with elevated levels of intracellular reactive oxygen species (ROS). ROS have been implicated in processes that promote viability, cell growth, and regulation of other biological functions such as migration of cells or gene expression. Currently, the BCR-ABL inhibitor imatinib mesylate (Gleevec) is being used as a first-line therapy for the treatment of CML. However, BCR-ABL transformation is associated with genomic instability, and disease progression or resistance to imatinib can occur. Imatinib resistance is not known to cause or significantly alter signaling requirements in transformed cells. Elevated ROS are crucial for transformation, making them an ideal additional target for therapeutic intervention. The underlying mechanisms leading to elevated oxidative stress are reviewed, and signaling mechanisms that may serve as novel targeted approaches to overcome ROS-dependent cell growth are discussed.
Collapse
Affiliation(s)
- Margret S Rodrigues
- Department of Medical Oncology, Dana-Farber Cancer Institute, Brigham and Women's Hospital, and Harvard Medical School, Boston, Massachusetts 02115, USA
| | | | | |
Collapse
|
27
|
Park H, Bahn YJ, Jung SK, Jeong DG, Lee SH, Seo I, Yoon TS, Kim SJ, Ryu SE. Discovery of novel Cdc25 phosphatase inhibitors with micromolar activity based on the structure-based virtual screening. J Med Chem 2008; 51:5533-41. [PMID: 18714978 DOI: 10.1021/jm701157g] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Cdc25 phosphatases have been considered as attractive drug targets for anticancer therapy because of the correlation of their overexpression with a wide variety of cancers. We have been able to identify five novel Cdc25 phosphatase inhibitors with micromolar activity by means of a computer-aided drug design protocol involving the homology modeling of Cdc25A and the virtual screening with the automated AutoDock program implementing the effects of ligand solvation in the scoring function. Because the newly discovered inhibitors are structurally diverse and reveal a significant potency with IC 50 values lower than 10 microM, they can be considered for further development by structure-activity relationship studies or de novo design methods. The differences in binding modes of the identified inhibitors in the active sites of Cdc25A and B are discussed in detail.
Collapse
Affiliation(s)
- Hwangseo Park
- Department of Bioscience and Biotechnology, Sejong University, 98 Kunja-Dong, Kwangjin-Ku, Seoul 143-747, Korea.
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Nanosecond molecular dynamics simulations of Cdc25B and its complex with a 1,4-naphthoquinone inhibitor: Implications for rational inhibitor design. J Mol Graph Model 2008; 27:13-9. [DOI: 10.1016/j.jmgm.2008.02.002] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2007] [Revised: 01/28/2008] [Accepted: 02/05/2008] [Indexed: 01/24/2023]
|
29
|
Toward the virtual screening of Cdc25A phosphatase inhibitors with the homology modeled protein structure. J Mol Model 2008; 14:833-41. [PMID: 18504625 DOI: 10.1007/s00894-008-0311-2] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/22/2007] [Accepted: 04/04/2008] [Indexed: 10/22/2022]
Abstract
Cdc25 phosphatases have been considered as attractive drug targets for anticancer therapy due to the correlation of their overexpression with a wide variety of cancers. As a method for the discovery of novel inhibitors of Cdc25 phosphatases, we have evaluated the computer-aided drug design protocol involving the homology modeling of Cdc25A and virtual screening with the two docking tools: FlexX and the modified AutoDock program implementing the effects of ligand solvation in the scoring function. The homology modeling with the X-ray crystal structure of Cdc25B as a template provides a high-quality structure of Cdc25A that enables the structure-based inhibitor design. Of the two docking programs under consideration, AutoDock is found to be more accurate than FlexX in terms of scoring putative ligands. A detailed binding mode analysis of the known inhibitors shows that they can be stabilized in the active site of Cdc25A through the simultaneous establishment of the multiple hydrogen bonds and the hydrophobic interactions. The present study demonstrates the usefulness of the modified AutoDock program as a docking tool for virtual screening of new Cdc25 phosphatase inhibitors as well as for binding mode analysis to elucidate the activities of known inhibitors.
Collapse
|
30
|
Didier C, Cavelier C, Quaranta M, Galcera MO, Demur C, Laurent G, Manenti S, Ducommun B. G2/M checkpoint stringency is a key parameter in the sensitivity of AML cells to genotoxic stress. Oncogene 2008; 27:3811-20. [PMID: 18212737 DOI: 10.1038/sj.onc.1211041] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Acute myeloid leukemia (AML) cells exposed to genotoxic agents arrest their cell cycle at the G2/M checkpoint and are inherently chemoresistant. To understand the mechanism of this chemoresistance, we compared the ability of immature CD34+ versus mature CD34- AML cell lines (KG1a and U937, respectively) to recover from a DNA damage-induced cell cycle checkpoint in G2. Here, we report that KG1a cells have a more stringent G2/M checkpoint response than U937 cells. We show that in both cell types, the CDC25B phosphatase participates in the G2/M checkpoint recovery and that its expression is upregulated. Furthermore, we show that CHK1 inhibition by UCN-01 in immature KG1a cells allows checkpoint exit and induces sensitivity to genotoxic agents. Similarly, UCN-01 treatment potentializes genotoxic-induced inhibition of colony formation efficiency of primary leukemic cells from AML patients. Altogether, our results demonstrate that checkpoint stringency varies during the maturation process and indicate that targeting checkpoint mechanisms might represent an attractive therapeutic opportunity for chemoresistant immature AML cells.
Collapse
Affiliation(s)
- C Didier
- LBCMCP-CNRS UMR5088-IFR109 Institut d'Exploration Fonctionnelle des Génomes, University of Toulouse, Toulouse, France
| | | | | | | | | | | | | | | |
Collapse
|
31
|
McLornan DP, McMullin MF, Johnston P, Longley DB. Molecular mechanisms of drug resistance in acute myeloid leukaemia. Expert Opin Drug Metab Toxicol 2007; 3:363-77. [PMID: 17539744 DOI: 10.1517/17425255.3.3.363] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
Resistance to chemotherapy in acute myeloid leukaemia is a major obstacle to a successful outcome for many patients. Often, there is resistance against a broad range of drugs due to multiple, simultaneously active processes. These mechanisms include effects on drug influx and efflux, drug activation/inactivation, DNA repair mechanisms, altered response of end targets, an altered haematopoietic microenvironment and dysfunctional apoptotic pathways. This article reviews the factors that determine leukaemic cell chemosensitivity and discusses the potential for rationally guided therapy.
Collapse
Affiliation(s)
- Donal P McLornan
- Medical Research Council Clinical Research Fellow, Queen's University Belfast, Centre for Cancer Research and Cell Biology, BT7 1NN, Northern Ireland, UK
| | | | | | | |
Collapse
|
32
|
Contour-Galcera MO, Sidhu A, Prévost G, Bigg D, Ducommun B. What's new on CDC25 phosphatase inhibitors. Pharmacol Ther 2007; 115:1-12. [PMID: 17531323 DOI: 10.1016/j.pharmthera.2007.03.009] [Citation(s) in RCA: 61] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2007] [Accepted: 03/26/2007] [Indexed: 11/30/2022]
Abstract
The CDC25 phosphatases are key regulators of cell cycle progression and play a central role in the checkpoint response to DNA damage. Their inhibition may therefore represent a promising therapeutic approach in oncology, and small molecule design strategies are currently leading to the identification of various classes of CDC25 inhibitors. Most structures developed so far are quinonoid-based compounds, but also phosphate surrogates or electrophilic entities. Considering the characteristics of the highly conserved active sites of the enzymes, many mechanisms of action have been proposed for these inhibitors. Quinonoid compounds may oxidize the catalytic site cysteine, but can also be considered as Michaël acceptors capable of reacting with the activated thiolate or other electrophilic entities. Phosphate surrogates are thought to interfere with the arginine residue, leading to reversible enzyme inhibition. But some inhibitors can combine in the same molecule several of these mechanisms, thus by fitting into the active site of the enzyme through one part of the molecule and bringing the reactive moiety in close proximity to the catalytic cysteine. This review summarizes novel classes of inhibitors that show specificity for the CDC25s over other phosphatases, cause cell proliferation inhibition and cell cycle arrest in vitro but also, for several of them, inhibition of xenografted tumoral cell growth in vivo. These promising results confirm the interest of the inhibition of CDC25 phosphatases as an anticancer therapeutic strategy.
Collapse
|