1
|
Choi K, Lee J, Kim G, Lim Y, Kang HJ. Recovery of synaptic loss and depressive-like behavior induced by GATA1 through blocking of the neuroinflammatory response. Front Cell Neurosci 2024; 18:1369951. [PMID: 38784708 PMCID: PMC11112091 DOI: 10.3389/fncel.2024.1369951] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/13/2024] [Accepted: 04/09/2024] [Indexed: 05/25/2024] Open
Abstract
GATA1, a member of the GATA transcription factor family, is a critical factor in hematopoietic system development. In a previous study, we demonstrated the increased expression of GATA1 in the dorsolateral prefrontal cortex (dlPFC) of patients suffering from depression and described its role as a transcriptional repressor of synapse-related genes. In this study, we investigated how GATA1 globally altered gene expression using multi-omics approaches. Through the combined analyses of ChIPseq, mRNAseq, and small RNAseq, we profiled genes that are potentially affected by GATA1 in cultured cortical neurons, and Gene Ontology (GO) analysis revealed that GATA1 might be associated with immune-related functions. We hypothesized that GATA1 induces immune activation, which has detrimental effects including synapse loss and depressive-like behavior. To test this hypothesis, we first performed a microglial morphometric analysis of a brain having overexpression of GATA1 because microglia are the resident immune cells of the central nervous system. Fractal analysis showed that the ramification and process length of microglia decreased in brains having GATA1 overexpression compared to the control, suggesting that GATA1 overexpression increases the activation of microglia. Through flow cytometry and immunohistochemical analysis, we found that activated microglia showed pro-inflammatory phenotypes characterized by the expression of CD86 and CD68. Finally, we demonstrated that the effects of GATA1 overexpression including synapse loss and depressive-like behavior could be blocked by inhibiting microglial activation using minocycline. These results will elucidate the regulatory mechanisms of GATA1 that affect pathophysiological conditions such as depression and provide a potential target for the treatment of depression.
Collapse
Affiliation(s)
| | | | | | | | - Hyo Jung Kang
- Department of Life Science, Chung-Ang University, Seoul, Republic of Korea
| |
Collapse
|
2
|
Li F, Xiong Y, Yang M, Chen P, Zhang J, Wang Q, Xu M, Wang Y, He Z, Zhao X, Huang J, Gu X, Zhang L, Sun R, Sun X, Li J, Ou J, Xu T, Huang X, Cao Y, Xu XR, Karakas D, Li J, Ni H, Zhang Q. c-Mpl-del, a c-Mpl alternative splicing isoform, promotes AMKL progression and chemoresistance. Cell Death Dis 2022; 13:869. [PMID: 36229456 PMCID: PMC9561678 DOI: 10.1038/s41419-022-05315-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2022] [Revised: 09/26/2022] [Accepted: 09/30/2022] [Indexed: 11/05/2022]
Abstract
Acute megakaryocytic leukemia (AMKL) is a clinically heterogeneous subtype of acute myeloid leukemia characterized by unrestricted megakaryoblast proliferation and poor prognosis. Thrombopoietin receptor c-Mpl is a primary regulator of megakaryopoeisis and a potent mitogenic receptor. Aberrant c-Mpl signaling has been implicated in a myriad of myeloid proliferative disorders, some of which can lead to AMKL, however, the role of c-Mpl in AMKL progression remains largely unexplored. Here, we identified increased expression of a c-Mpl alternative splicing isoform, c-Mpl-del, in AMKL patients. We found that c-Mpl-del expression was associated with enhanced AMKL cell proliferation and chemoresistance, and decreased survival in xenografted mice, while c-Mpl-del knockdown attenuated proliferation and restored apoptosis. Interestingly, we observed that c-Mpl-del exhibits preferential utilization of phosphorylated c-Mpl-del C-terminus Y607 and biased activation of PI3K/AKT pathway, which culminated in upregulation of GATA1 and downregulation of DDIT3-related apoptotic responses conducive to AMKL chemoresistance and proliferation. Thus, this study elucidates the critical roles of c-Mpl alternative splicing in AMKL progression and drug resistance, which may have important diagnostic and therapeutic implications for leukemia accelerated by c-Mpl-del overexpression.
Collapse
Affiliation(s)
- Fei Li
- grid.12981.330000 0001 2360 039XState Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yuanyan Xiong
- grid.12981.330000 0001 2360 039XState Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Mo Yang
- grid.12981.330000 0001 2360 039XThe Seventh Affiliated Hospital, Sun Yat-sen University, Shenzhen, China
| | - Peiling Chen
- grid.12981.330000 0001 2360 039XState Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jingkai Zhang
- grid.12981.330000 0001 2360 039XState Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Qiong Wang
- grid.12981.330000 0001 2360 039XState Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China ,grid.12981.330000 0001 2360 039XInstitute of Sun Yat-sen University in Shenzhen, Shenzhen, China
| | - Miao Xu
- grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, and Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Canada
| | - Yiming Wang
- grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, and Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Canada ,Canadian Blood Services Centre for Innovation, Toronto, Canada
| | - Zuyong He
- grid.12981.330000 0001 2360 039XState Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xin Zhao
- grid.12981.330000 0001 2360 039XState Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Junyu Huang
- grid.12981.330000 0001 2360 039XState Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiaoqiong Gu
- grid.410737.60000 0000 8653 1072Department of Blood Transfusion, Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Li Zhang
- grid.410737.60000 0000 8653 1072Department of Blood Transfusion, Clinical Biological Resource Bank and Clinical Lab, Guangzhou Institute of Pediatrics, Guangzhou Women and Children’s Medical Center, Guangzhou Medical University, Guangzhou, China
| | - Rui Sun
- grid.488530.20000 0004 1803 6191State Key Laboratory of Oncology in South China, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xunsha Sun
- grid.12981.330000 0001 2360 039XNational Key Clinical Department and Key Discipline of Neurology, The First Affiliated Hospital, Sun Yat-sen University, Guangzhou, China
| | - Jingyao Li
- grid.12981.330000 0001 2360 039XState Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Jinxin Ou
- grid.12981.330000 0001 2360 039XState Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Ting Xu
- grid.12981.330000 0001 2360 039XState Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xueying Huang
- grid.12981.330000 0001 2360 039XState Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Yange Cao
- grid.12981.330000 0001 2360 039XState Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China
| | - Xiaohong Ruby Xu
- grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, and Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Canada
| | - Danielle Karakas
- grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, and Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Canada
| | - June Li
- grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, and Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Canada ,Canadian Blood Services Centre for Innovation, Toronto, Canada
| | - Heyu Ni
- grid.17063.330000 0001 2157 2938Department of Laboratory Medicine and Pathobiology, University of Toronto, Toronto, Canada ,grid.17063.330000 0001 2157 2938Department of Laboratory Medicine, Keenan Research Centre for Biomedical Science of St. Michael’s Hospital, and Toronto Platelet Immunobiology Group, University of Toronto, Toronto, Canada ,Canadian Blood Services Centre for Innovation, Toronto, Canada ,grid.17063.330000 0001 2157 2938Department of Physiology, University of Toronto, Toronto, Canada
| | - Qing Zhang
- grid.12981.330000 0001 2360 039XState Key Laboratory of Biocontrol, School of Life Sciences, Sun Yat-sen University, Guangzhou, China ,grid.12981.330000 0001 2360 039XInstitute of Sun Yat-sen University in Shenzhen, Shenzhen, China
| |
Collapse
|
3
|
Repurposing of the ALK Inhibitor Crizotinib for Acute Leukemia and Multiple Myeloma Cells. Pharmaceuticals (Basel) 2021; 14:ph14111126. [PMID: 34832908 PMCID: PMC8617756 DOI: 10.3390/ph14111126] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/14/2021] [Revised: 10/28/2021] [Accepted: 11/02/2021] [Indexed: 12/18/2022] Open
Abstract
Crizotinib was a first generation of ALK tyrosine kinase inhibitor approved for the treatment of ALK-positive non-small-cell lung carcinoma (NSCLC) patients. COMPARE and cluster analyses of transcriptomic data of the NCI cell line panel indicated that genes with different cellular functions regulated the sensitivity or resistance of cancer cells to crizotinib. Transcription factor binding motif analyses in gene promoters divulged two transcription factors possibly regulating the expression of these genes, i.e., RXRA and GATA1, which are important for leukemia and erythroid development, respectively. COMPARE analyses also implied that cell lines of various cancer types displayed varying degrees of sensitivity to crizotinib. Unexpectedly, leukemia but not lung cancer cells were the most sensitive cells among the different types of NCI cancer cell lines. Re-examining this result in another panel of cell lines indeed revealed that crizotinib exhibited potent cytotoxicity towards acute myeloid leukemia and multiple myeloma cells. P-glycoprotein-overexpressing CEM/ADR5000 leukemia cells were cross-resistant to crizotinib. NCI-H929 multiple myeloma cells were the most sensitive cells. Hence, we evaluated the mode of action of crizotinib on these cells. Although crizotinib is a TKI, it showed highest correlation rates with DNA topoisomerase II inhibitors and tubulin inhibitors. The altered gene expression profiles after crizotinib treatment predicted several networks, where TOP2A and genes related to cell cycle were downregulated. Cell cycle analyses showed that cells incubated with crizotinib for 24 h accumulated in the G2M phase. Crizotinib also increased the number of p-H3(Ser10)-positive NCI-H929 cells illustrating crizotinib's ability to prevent mitotic exit. However, cells accumulated in the sub-G0G1 fraction with longer incubation periods, indicating apoptosis induction. Additionally, crizotinib disassembled the tubulin network of U2OS cells expressing an α-tubulin-GFP fusion protein, preventing migration of cancer cells. This result was verified by in vitro tubulin polymerization assays. In silico molecular docking also revealed a strong binding affinity of crizotinib to the colchicine and Vinca alkaloid binding sites. Taken together, these results demonstrate that crizotinib destabilized microtubules. Additionally, the decatenation assay showed that crizotinib partwise inhibited the catalytic activity of DNA topoisomerase II. In conclusion, crizotinib exerted kinase-independent cytotoxic effects through the dual inhibition of tubulin polymerization and topoisomerase II and might be used to treat not only NSCLC but also multiple myeloma.
Collapse
|
4
|
Chung HY, Lin BA, Lin YX, Chang CW, Tzou WS, Pei TW, Hu CH. Meis1, Hi1α, and GATA1 are integrated into a hierarchical regulatory network to mediate primitive erythropoiesis. FASEB J 2021; 35:e21915. [PMID: 34496088 DOI: 10.1096/fj.202001044rrr] [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: 04/29/2020] [Revised: 08/20/2021] [Accepted: 08/27/2021] [Indexed: 12/16/2022]
Abstract
During development, erythroid cells are generated by two waves of hematopoiesis. In zebrafish, primitive erythropoiesis takes place in the intermediate cell mass region, and definitive erythropoiesis arises from the aorta-gonad mesonephros. TALE-homeoproteins Meis1 and Pbx1 function upstream of GATA1 to specify the erythroid lineage. Embryos lacking Meis1 or Pbx1 have weak gata1 expression and fail to produce primitive erythrocytes. Nevertheless, the underlying mechanism of how Meis1 and Pbx1 mediate gata1 transcription in erythrocytes remains unclear. Here we show that Hif1α acts downstream of Meis1 to mediate gata1 expression in zebrafish embryos. Inhibition of Meis1 expression resulted in suppression of hif1a expression and abrogated primitive erythropoiesis, while injection with in vitro-synthesized hif1α mRNA rescued gata1 transcription in Meis1 morphants and recovered their erythropoiesis. Ablation of Hif1α expression either by morpholino knockdown or Crispr-Cas9 knockout suppressed gata1 transcription and abrogated primitive erythropoiesis. Results of chromatin immunoprecipitation assays showed that Hif1α associates with hypoxia-response elements located in the 3'-flanking region of gata1 during development, suggesting that Hif1α regulates gata1 expression in vivo. Together, our results indicate that Meis1, Hif1α, and GATA1 indeed comprise a hierarchical regulatory network in which Hif1α acts downstream of Meis1 to activate gata1 transcription through direct interactions with its cis-acting elements in primitive erythrocytes.
Collapse
Affiliation(s)
- Hsin-Yu Chung
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, Taiwan
| | - Bo-An Lin
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, Taiwan
| | - Yi-Xuan Lin
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, Taiwan
| | - Chen-Wei Chang
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, Taiwan
| | - Wen-Shyong Tzou
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, Taiwan.,Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung, Taiwan
| | - Tun-Wen Pei
- Department of Computer Science and Information Engineering, National Taipei University of Technology
| | - Chin-Hwa Hu
- Department of Bioscience and Biotechnology, National Taiwan Ocean University, Keelung, Taiwan.,Center of Excellence for the Oceans, National Taiwan Ocean University, Keelung, Taiwan
| |
Collapse
|
5
|
Xie J, Zhao C, Sun J, Li J, Yang F, Wang J, Nie Q. Prediction of Essential Genes in Comparison States Using Machine Learning. IEEE/ACM TRANSACTIONS ON COMPUTATIONAL BIOLOGY AND BIOINFORMATICS 2021; 18:1784-1792. [PMID: 32991286 DOI: 10.1109/tcbb.2020.3027392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 06/11/2023]
Abstract
Identifying essential genes in comparison states (EGS) is vital to understanding cell differentiation, performing drug discovery, and identifying disease causes. Here, we present a machine learning method termed Prediction of Essential Genes in Comparison States (PreEGS). To capture the alteration of the network in comparison states, PreEGS extracts topological and gene expression features of each gene in a five-dimensional vector. PreEGS also recruits a positive sample expansion method to address the problem of unbalanced positive and negative samples, which is often encountered in practical applications. Different classifiers are applied to the simulated datasets, and the PreEGS based on the random forests model (PreEGSRF) was chosen for optimal performance. PreEGSRF was then compared with six other methods, including three machine learning methods, to predict EGS in a specific state. On real datasets with four gene regulatory networks, PreEGSRF predicted five essential genes related to leukemia and five enriched KEGG pathways. Four of the predicted essential genes and all predicted pathways were consistent with previous studies and highly correlated with leukemia. With high prediction accuracy and generalization ability, PreEGSRF is broadly applicable for the discovery of disease-causing genes, driver genes for cell fate decisions, and complex biomarkers of biological systems.
Collapse
|
6
|
Creed TM, Baldeosingh R, Eberly CL, Schlee CS, Kim M, Cutler JA, Pandey A, Civin CI, Fossett NG, Kingsbury TJ. The PAX-SIX-EYA-DACH network modulates GATA-FOG function in fly hematopoiesis and human erythropoiesis. Development 2020; 147:dev.177022. [PMID: 31806659 DOI: 10.1242/dev.177022] [Citation(s) in RCA: 4] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/19/2019] [Accepted: 11/25/2019] [Indexed: 12/15/2022]
Abstract
The GATA and PAX-SIX-EYA-DACH transcriptional networks (PSEDNs) are essential for proper development across taxa. Here, we demonstrate novel PSEDN roles in vivo in Drosophila hematopoiesis and in human erythropoiesis in vitro Using Drosophila genetics, we show that PSEDN members function with GATA to block lamellocyte differentiation and maintain the prohemocyte pool. Overexpression of human SIX1 stimulated erythroid differentiation of human erythroleukemia TF1 cells and primary hematopoietic stem-progenitor cells. Conversely, SIX1 knockout impaired erythropoiesis in both cell types. SIX1 stimulation of erythropoiesis required GATA1, as SIX1 overexpression failed to drive erythroid phenotypes and gene expression patterns in GATA1 knockout cells. SIX1 can associate with GATA1 and stimulate GATA1-mediated gene transcription, suggesting that SIX1-GATA1 physical interactions contribute to the observed functional interactions. In addition, both fly and human SIX proteins regulated GATA protein levels. Collectively, our findings demonstrate that SIX proteins enhance GATA function at multiple levels, and reveal evolutionarily conserved cooperation between the GATA and PSEDN networks that may regulate developmental processes beyond hematopoiesis.
Collapse
Affiliation(s)
- T Michael Creed
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Rajkumar Baldeosingh
- Center for Vascular and Inflammatory Diseases University of Maryland School of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Manipal Academy of Higher Education (MAHE), Manipal 576104, Karnataka, India
| | - Christian L Eberly
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Caroline S Schlee
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - MinJung Kim
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Jevon A Cutler
- McKusick-Nathans Institute of Genetic Medicine, Departments of Biological Chemistry, Oncology and Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Akhilesh Pandey
- McKusick-Nathans Institute of Genetic Medicine, Departments of Biological Chemistry, Oncology and Pathology, Johns Hopkins University School of Medicine, Baltimore, MD 21205, USA
| | - Curt I Civin
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Department of Pediatrics, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Nancy G Fossett
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA .,Center for Vascular and Inflammatory Diseases University of Maryland School of Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| | - Tami J Kingsbury
- Center for Stem Cell Biology & Regenerative Medicine, University of Maryland School of Medicine, Baltimore, MD 21201, USA .,Marlene and Stewart Greenebaum Comprehensive Cancer Center, University of Maryland School of Medicine, Baltimore, MD 21201, USA.,Department of Physiology, University of Maryland School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
7
|
De Marchi F, Araki M, Komatsu N. Molecular features, prognosis, and novel treatment options for pediatric acute megakaryoblastic leukemia. Expert Rev Hematol 2019; 12:285-293. [PMID: 30991862 DOI: 10.1080/17474086.2019.1609351] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
INTRODUCTION Acute megakaryoblastic leukemia (AMegL) is a rare hematological neoplasm most often diagnosed in children and is commonly associated with Down's syndrome (DS). Although AMegLs are specifically characterized and typically diagnosed by megakaryoblastic expansion, recent advancements in molecular analysis have highlighted the heterogeneity of this disease, with specific cytogenic and genetic alterations characterizing different disease subtypes. Areas covered: This review will focus on describing recurrent molecular variations in both DS and non-DS pediatric AMegL, their role in promoting leukemogenesis, their association with different clinical aspects and prognosis, and finally, their influence on future treatment strategies with a number of specific drugs beyond conventional chemotherapy already under development. Expert opinion: Deep understanding of the genetic and molecular landscape of AMegL will lead to better and more precise disease classification in terms of diagnosis, prognosis, and possible targeted therapies. Development of new therapeutic approaches based on these molecular characteristics will hopefully improve AMegL patient outcomes.
Collapse
Affiliation(s)
- Federico De Marchi
- a Department of Hematology , Juntendo University Graduate School of Medicine , Tokyo , Japan
| | - Marito Araki
- b Department of Transfusion Medicine and Stem Cell Regulation , Juntendo University Graduate School of Medicine , Tokyo , Japan
| | - Norio Komatsu
- a Department of Hematology , Juntendo University Graduate School of Medicine , Tokyo , Japan
| |
Collapse
|
8
|
Garcia-Carpizo V, Ruiz-Llorente S, Sarmentero J, Graña-Castro O, Pisano DG, Barrero MJ. CREBBP/EP300 bromodomains are critical to sustain the GATA1/MYC regulatory axis in proliferation. Epigenetics Chromatin 2018; 11:30. [PMID: 29884215 PMCID: PMC5992658 DOI: 10.1186/s13072-018-0197-x] [Citation(s) in RCA: 37] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/24/2018] [Accepted: 05/27/2018] [Indexed: 02/06/2023] Open
Abstract
Background The reported antitumor activity of the BET family bromodomain inhibitors has prompted the development of inhibitors against other bromodomains. However, the human genome encodes more than 60 different bromodomains and most of them remain unexplored. Results We report that the bromodomains of the histone acetyltransferases CREBBP/EP300 are critical to sustain the proliferation of human leukemia and lymphoma cell lines. EP300 is very abundant at super-enhancers in K562 and is coincident with sites of GATA1 and MYC occupancy. In accordance, CREBBP/EP300 bromodomain inhibitors interfere with GATA1- and MYC-driven transcription, causing the accumulation of cells in the G0/G1 phase of the cell cycle. The CREBBP/CBP30 bromodomain inhibitor CBP30 displaces CREBBP and EP300 from GATA1 and MYC binding sites at enhancers, resulting in a decrease in the levels of histone acetylation at these regulatory regions and consequently reduced gene expression of critical genes controlled by these transcription factors. Conclusions Our data shows that inhibition of CREBBP/EP300 bromodomains can interfere with oncogene-driven transcriptional programs in cancer cells and consequently hold therapeutic potential. Electronic supplementary material The online version of this article (10.1186/s13072-018-0197-x) contains supplementary material, which is available to authorized users.
Collapse
Affiliation(s)
- Veronica Garcia-Carpizo
- CNIO-Lilly Epigenetics Laboratory, Spanish National Cancer Research Center (CNIO), Melchor Fernandez Almagro 3, 28029, Madrid, Spain
| | - Sergio Ruiz-Llorente
- CNIO-Lilly Epigenetics Laboratory, Spanish National Cancer Research Center (CNIO), Melchor Fernandez Almagro 3, 28029, Madrid, Spain
| | - Jacinto Sarmentero
- CNIO-Lilly Epigenetics Laboratory, Spanish National Cancer Research Center (CNIO), Melchor Fernandez Almagro 3, 28029, Madrid, Spain
| | - Osvaldo Graña-Castro
- Bioinformatics Unit, Spanish National Cancer Research Center (CNIO), Melchor Fernandez Almagro 3, 28029, Madrid, Spain
| | - David G Pisano
- Bioinformatics Unit, Spanish National Cancer Research Center (CNIO), Melchor Fernandez Almagro 3, 28029, Madrid, Spain
| | - Maria J Barrero
- CNIO-Lilly Epigenetics Laboratory, Spanish National Cancer Research Center (CNIO), Melchor Fernandez Almagro 3, 28029, Madrid, Spain.
| |
Collapse
|
9
|
Scheenstra MR, De Cuyper IM, Branco-Madeira F, de Bleser P, Kool M, Meinders M, Hoogenboezem M, Mul E, Wolkers MC, Salerno F, Nota B, Saeys Y, Klarenbeek S, van IJcken WFJ, Hammad H, Philipsen S, van den Berg TK, Kuijpers TW, Lambrecht BN, Gutiérrez L. GATA1-Deficient Dendritic Cells Display Impaired CCL21-Dependent Migration toward Lymph Nodes Due to Reduced Levels of Polysialic Acid. THE JOURNAL OF IMMUNOLOGY 2016; 197:4312-4324. [DOI: 10.4049/jimmunol.1600103] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/21/2016] [Accepted: 09/29/2016] [Indexed: 02/05/2023]
|
10
|
Acute megakaryocytic leukemia: What have we learned. Blood Rev 2016; 30:49-53. [DOI: 10.1016/j.blre.2015.07.005] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2015] [Revised: 06/04/2015] [Accepted: 07/10/2015] [Indexed: 11/23/2022]
|
11
|
Scheenstra MR, Salunkhe V, De Cuyper IM, Hoogenboezem M, Li E, Kuijpers TW, van den Berg TK, Gutiérrez L. Characterization of hematopoietic GATA transcription factor expression in mouse and human dendritic cells. Blood Cells Mol Dis 2015; 55:293-303. [PMID: 26460250 DOI: 10.1016/j.bcmd.2015.07.006] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2015] [Revised: 07/10/2015] [Accepted: 07/11/2015] [Indexed: 12/24/2022]
Abstract
Dendritic cells (DCs) are key initiators and regulators of the immune response. The development of the DC lineage and their subsets requires an orchestrated regulation of their transcriptional program. Gata1, a transcription factor expressed in several hematopoietic cell lineages, has been recently reported to be required for mouse DC development and function. In humans, GATA1 is involved in the lineage separation between monocyte-derived DCs and Langerhans cells (LC) and loss of GATA1 results in differentiation arrest at the monocyte stage. The hematopoietic GATA factors (i.e. Gata1, Gata2, Gata3) are known to regulate each other's expression and to function consecutively throughout lineage commitment (so-called GATA switch). In humans, mutations in GATA2 are causative of MonoMAC disease, a human immunodeficiency syndrome characterized by loss of DCs, monocytes, B and NK cells. However, additional data on the expression of hematopoietic GATA factors in the DC lineage is missing. In this study, we have characterized the expression of hematopoietic GATA factors in murine and human DCs and their expression dynamics upon TLR stimulation. We found that all hematopoietic GATA factors are expressed in DCs, but identified species-specific differences in the relative expression of each GATA factor, and how their expression fluctuates upon stimulation.
Collapse
Affiliation(s)
- Maaike R Scheenstra
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Vishal Salunkhe
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Iris M De Cuyper
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Mark Hoogenboezem
- Department of Molecular Cell Biology, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Eveline Li
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Taco W Kuijpers
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands; Department of Pediatric Hematology, Immunology and Infectious Diseases, Emma Children's Hospital, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Timo K van den Berg
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands
| | - Laura Gutiérrez
- Department of Blood Cell Research, Sanquin Research and Landsteiner Laboratory, Academic Medical Center, University of Amsterdam, Amsterdam, The Netherlands.
| |
Collapse
|
12
|
Mateos MK, Barbaric D, Byatt SA, Sutton R, Marshall GM. Down syndrome and leukemia: insights into leukemogenesis and translational targets. Transl Pediatr 2015; 4:76-92. [PMID: 26835364 PMCID: PMC4729084 DOI: 10.3978/j.issn.2224-4336.2015.03.03] [Citation(s) in RCA: 32] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Abstract
Children with Down syndrome (DS) have a significantly increased risk of childhood leukemia, in particular acute megakaryoblastic leukemia (AMKL) and acute lymphoblastic leukemia (DS-ALL). A pre-leukemia, called transient myeloproliferative disorder (TMD), characterised by a GATA binding protein 1 (GATA1) mutation, affects up to 30% of newborns with DS. In most cases, the pre-leukemia regresses spontaneously, however one-quarter of these children will go on to develop AMKL or myelodysplastic syndrome (MDS) . AMKL and MDS occurring in young children with DS and a GATA1 somatic mutation are collectively termed myeloid leukemia of Down syndrome (ML-DS). This model represents an important multi-step process of leukemogenesis, and further study is required to identify therapeutic targets to potentially prevent development of leukemia. DS-ALL is a high-risk leukemia and mutations in the JAK-STAT pathway are frequently observed. JAK inhibitors may improve outcome for this type of leukemia. Genetic and epigenetic studies have revealed likely candidate drivers involved in development of ML-DS and DS-ALL. Overall this review aims to identify potential impacts of new research on how we manage children with DS, pre-leukemia and leukemia.
Collapse
Affiliation(s)
- Marion K Mateos
- 1 Kids Cancer Centre, Sydney Children's Hospital, Randwick, Australia ; 2 School of Women's and Children's Health, University of New South Wales, Kensington, Australia ; 3 Children's Cancer Institute Australia, University of New South Wales, Lowy Cancer Centre, Randwick, Australia
| | - Draga Barbaric
- 1 Kids Cancer Centre, Sydney Children's Hospital, Randwick, Australia ; 2 School of Women's and Children's Health, University of New South Wales, Kensington, Australia ; 3 Children's Cancer Institute Australia, University of New South Wales, Lowy Cancer Centre, Randwick, Australia
| | - Sally-Anne Byatt
- 1 Kids Cancer Centre, Sydney Children's Hospital, Randwick, Australia ; 2 School of Women's and Children's Health, University of New South Wales, Kensington, Australia ; 3 Children's Cancer Institute Australia, University of New South Wales, Lowy Cancer Centre, Randwick, Australia
| | - Rosemary Sutton
- 1 Kids Cancer Centre, Sydney Children's Hospital, Randwick, Australia ; 2 School of Women's and Children's Health, University of New South Wales, Kensington, Australia ; 3 Children's Cancer Institute Australia, University of New South Wales, Lowy Cancer Centre, Randwick, Australia
| | - Glenn M Marshall
- 1 Kids Cancer Centre, Sydney Children's Hospital, Randwick, Australia ; 2 School of Women's and Children's Health, University of New South Wales, Kensington, Australia ; 3 Children's Cancer Institute Australia, University of New South Wales, Lowy Cancer Centre, Randwick, Australia
| |
Collapse
|
13
|
Mir MA, Kochuparambil ST, Abraham RS, Rodriguez V, Howard M, Hsu AP, Jackson AE, Holland SM, Patnaik MM. Spectrum of myeloid neoplasms and immune deficiency associated with germline GATA2 mutations. Cancer Med 2015; 4:490-9. [PMID: 25619630 PMCID: PMC4402062 DOI: 10.1002/cam4.384] [Citation(s) in RCA: 41] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/24/2014] [Revised: 10/21/2014] [Accepted: 11/02/2014] [Indexed: 01/25/2023] Open
Abstract
Guanine-adenine-thymine-adenine 2 (GATA2) mutated disorders include the recently described MonoMAC syndrome (Monocytopenia and Mycobacterium avium complex infections), DCML (dendritic cell, monocyte, and lymphocyte deficiency), familial MDS/AML (myelodysplastic syndrome/acute myeloid leukemia) (myeloid neoplasms), congenital neutropenia, congenital lymphedema (Emberger's syndrome), sensorineural deafness, viral warts, and a spectrum of aggressive infections seen across all age groups. While considerable efforts have been made to identify the mutations that characterize this disorder, pathogenesis remains a work in progress with less than 100 patients described in current literature. Varying clinical presentations offer diagnostic challenges. Allogeneic stem cell transplant remains the treatment of choice. Morbidity, mortality, and social costs due to the familial nature of the disease are considerable. We describe our experience with the disorder in three affected families and a comprehensive review of current literature.
Collapse
Affiliation(s)
- Muhammad A Mir
- Penn State Milton S. Hershey Cancer Institute, Hershey, Pennsylvania
| | | | | | | | | | | | | | | | | |
Collapse
|
14
|
Liu Q, Su PF, Zhao S, Shyr Y. Transcriptome-wide signatures of tumor stage in kidney renal clear cell carcinoma: connecting copy number variation, methylation and transcription factor activity. Genome Med 2014; 6:117. [PMID: 25648588 PMCID: PMC4293006 DOI: 10.1186/s13073-014-0117-z] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/13/2014] [Accepted: 11/26/2014] [Indexed: 01/10/2023] Open
Abstract
BACKGROUND Comparative analysis of expression profiles between early and late stage cancers can help to understand cancer progression and metastasis mechanisms and to predict the clinical aggressiveness of cancer. The observed stage-dependent expression changes can be explained by genetic and epigenetic alterations as well as transcription dysregulation. Unlike genetic and epigenetic alterations, however, activity changes of transcription factors, generally occurring at the post-transcriptional or post-translational level, are hard to detect and quantify. METHODS Here we developed a statistical framework to infer the activity changes of transcription factors by simultaneously taking into account the contributions of genetic and epigenetic alterations to mRNA expression variations. RESULTS Applied to kidney renal clear cell carcinoma (KIRC), the model underscored the role of methylation as a significant contributor to stage-dependent expression alterations and identified key transcription factors as potential drivers of cancer progression. CONCLUSIONS Integrating copy number, methylation, and transcription factor activity signatures to explain stage-dependent expression alterations presented a precise and comprehensive view on the underlying mechanisms during KIRC progression.
Collapse
Affiliation(s)
- Qi Liu
- Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232 USA ; Department of Biomedical Informatics, Vanderbilt University School of Medicine, Nashville, TN 37232 USA
| | - Pei-Fang Su
- Department of Statistics, National Cheng Kung University, Tainan, 70101 Taiwan
| | - Shilin Zhao
- Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232 USA
| | - Yu Shyr
- Center for Quantitative Sciences, Vanderbilt University School of Medicine, Nashville, TN 37232 USA ; Department of Cancer Biology, Vanderbilt University School of Medicine, Nashville, TN 37232 USA ; Department of Biostatistics, Vanderbilt University School of Medicine, Nashville, TN 37232 USA ; School of Life Sciences & Biotechnology, Shanghai Jiao Tong University, Shanghai, 200240 China
| |
Collapse
|
15
|
Pelleri MC, Piovesan A, Caracausi M, Berardi AC, Vitale L, Strippoli P. Integrated differential transcriptome maps of Acute Megakaryoblastic Leukemia (AMKL) in children with or without Down Syndrome (DS). BMC Med Genomics 2014; 7:63. [PMID: 25476127 PMCID: PMC4304173 DOI: 10.1186/s12920-014-0063-z] [Citation(s) in RCA: 20] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2014] [Accepted: 11/12/2014] [Indexed: 12/31/2022] Open
Abstract
BACKGROUND The incidence of Acute Megakaryoblastic Leukemia (AMKL) is 500-fold higher in children with Down Syndrome (DS) compared with non-DS children, but the relevance of trisomy 21 as a specific background of AMKL in DS is still an open issue. Several Authors have determined gene expression profiles by microarray analysis in DS and/or non-DS AMKL. Due to the rarity of AMKL, these studies were typically limited to a small group of samples. METHODS We generated integrated quantitative transcriptome maps by systematic meta-analysis from any available gene expression profile dataset related to AMKL in pediatric age. This task has been accomplished using a tool recently described by us for the generation and the analysis of quantitative transcriptome maps, TRAM (Transcriptome Mapper), which allows effective integration of data obtained from different experimenters, experimental platforms and data sources. This allowed us to explore gene expression changes involved in transition from normal megakaryocytes (MK, n=19) to DS (n=43) or non-DS (n=45) AMKL blasts, including the analysis of Transient Myeloproliferative Disorder (TMD, n=20), a pre-leukemia condition. RESULTS We propose a biological model of the transcriptome depicting progressive changes from MK to TMD and then to DS AMKL. The data indicate the repression of genes involved in MK differentiation, in particular the cluster on chromosome 4 including PF4 (platelet factor 4) and PPBP (pro-platelet basic protein); the gene for the mitogen-activated protein kinase MAP3K10 and the thrombopoietin receptor gene MPL. Moreover, comparing both DS and non-DS AMKL with MK, we identified three potential clinical markers of progression to AMKL: TMEM241 (transmembrane protein 241) was the most over-expressed single gene, while APOC2 (apolipoprotein C-II) and ZNF587B (zinc finger protein 587B) appear to be the most discriminant markers of progression, specifically to DS AMKL. Finally, the chromosome 21 (chr21) genes resulted to be the most over-expressed in DS and non-DS AMKL, as well as in TMD, pointing out a key role of chr21 genes in differentiating AMKL from MK. CONCLUSIONS Our study presents an integrated original model of the DS AMLK transcriptome, providing the identification of genes relevant for its pathophysiology which can potentially be new clinical markers.
Collapse
Affiliation(s)
- Maria Chiara Pelleri
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Unit of Histology, Embryology and Applied Biology, University of Bologna, Via Belmeloro 8, 40126, Bologna, BO, Italy.
| | - Allison Piovesan
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Unit of Histology, Embryology and Applied Biology, University of Bologna, Via Belmeloro 8, 40126, Bologna, BO, Italy.
| | - Maria Caracausi
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Unit of Histology, Embryology and Applied Biology, University of Bologna, Via Belmeloro 8, 40126, Bologna, BO, Italy.
| | - Anna Concetta Berardi
- Research Laboratory Stem Cells, U.O.C. Immunohematology-Transfusion Medicine and Laboratory of Hematology, Santo Spirito's Hospital, Via del Circuito, 65100, Pescara, Italy.
| | - Lorenza Vitale
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Unit of Histology, Embryology and Applied Biology, University of Bologna, Via Belmeloro 8, 40126, Bologna, BO, Italy.
| | - Pierluigi Strippoli
- Department of Experimental, Diagnostic and Specialty Medicine (DIMES), Unit of Histology, Embryology and Applied Biology, University of Bologna, Via Belmeloro 8, 40126, Bologna, BO, Italy. .,Interdepartmental Center for Cancer Research Giorgio Prodi (CIRC), S. Orsola-Malpighi Hospital, University of Bologna, Via Massarenti 9, 40138, Bologna, BO, Italy.
| |
Collapse
|
16
|
Xie C, Edwards H, Caldwell JT, Wang G, Taub JW, Ge Y. Obatoclax potentiates the cytotoxic effect of cytarabine on acute myeloid leukemia cells by enhancing DNA damage. Mol Oncol 2014; 9:409-21. [PMID: 25308513 DOI: 10.1016/j.molonc.2014.09.008] [Citation(s) in RCA: 26] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/18/2014] [Revised: 09/09/2014] [Accepted: 09/16/2014] [Indexed: 12/13/2022] Open
Abstract
Resistance to cytarabine and anthracycline-based chemotherapy is a major cause of treatment failure for acute myeloid leukemia (AML) patients. Overexpression of Bcl-2, Bcl-xL, and/or Mcl-1 has been associated with chemoresistance in AML cell lines and with poor clinical outcome of AML patients. Thus, inhibitors of anti-apoptotic Bcl-2 family proteins could be novel therapeutic agents. In this study, we investigated how clinically achievable concentrations of obatoclax, a pan-Bcl-2 inhibitor, potentiate the antileukemic activity of cytarabine in AML cells. MTT assays in AML cell lines and diagnostic blasts, as well as flow cytometry analyses in AML cell lines revealed synergistic antileukemic activity between cytarabine and obatoclax. Bax activation was detected in the combined, but not the individual, drug treatments. This was accompanied by significantly increased loss of mitochondrial membrane potential. Most importantly, in AML cells treated with the combination, enhanced early induction of DNA double-strand breaks (DSBs) preceded a decrease of Mcl-1 levels, nuclear translocation of Bcl-2, Bcl-xL, and Mcl-1, and apoptosis. These results indicate that obatoclax enhances cytarabine-induced apoptosis by enhancing DNA DSBs. This novel mechanism provides compelling evidence for the clinical use of BH3 mimetics in combination with DNA-damaging agents in AML and possibly a broader range of malignancies.
Collapse
Affiliation(s)
- Chengzhi Xie
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA; National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, PR China
| | - Holly Edwards
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA
| | - J Timothy Caldwell
- MD/PhD Program, Wayne State University School of Medicine, Detroit, MI, USA; Cancer Biology Program, Wayne State University School of Medicine, Detroit, MI, USA
| | - Guan Wang
- National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, PR China; Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Jeffrey W Taub
- Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA; Division of Pediatric Hematology/Oncology, Children's Hospital of Michigan, Detroit, MI, USA; Department of Pediatrics, Wayne State University School of Medicine, Detroit, MI, USA
| | - Yubin Ge
- Department of Oncology, Wayne State University School of Medicine, Detroit, MI, USA; Molecular Therapeutics Program, Barbara Ann Karmanos Cancer Institute, Wayne State University School of Medicine, Detroit, MI, USA; National Engineering Laboratory for AIDS Vaccine, Key Laboratory for Molecular Enzymology and Engineering, The Ministry of Education, School of Life Sciences, Jilin University, Changchun, PR China.
| |
Collapse
|