171 results on '"Zhizhuang Joe Zhao"'
Search Results
2. miR-221/222 induce instability of p53 By downregulating deubiquitinase YOD1 in acute myeloid leukemia
- Author
-
Han Zhong Pei, Zhiyong Peng, Xiaomei Zhuang, Xiaobo Wang, Bo Lu, Yao Guo, Yuming Zhao, Dengyang Zhang, Yunjun Xiao, Tianshun Gao, Liuting Yu, Chunxiao He, Shunjie Wu, Suk-Hwan Baek, Zhizhuang Joe Zhao, Xiaojun Xu, and Yun Chen
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 ,Cytology ,QH573-671 - Abstract
Abstract Acute myeloid leukemia (AML) is a hematological malignancy characterized by the impaired differentiation and uncontrolled proliferation of myeloid blasts. Tumor suppressor p53 is often downregulated in AML cells via ubiquitination-mediated degradation. While the role of E3 ligase MDM2 in p53 ubiquitination is well-accepted, little is known about the involvement of deubiquitinases (DUBs). Herein, we found that the expression of YOD1, among several DUBs, is substantially reduced in blood cells from AML patients. We identified that YOD1 deubiqutinated and stabilized p53 through interaction via N-terminus of p53 and OTU domain of YOD1. In addition, expression levels of YOD1 were suppressed by elevated miR-221/222 in AML cells through binding to the 3′ untranslated region of YOD1, as verified by reporter gene assays. Treatment of cells with miR-221/222 mimics and inhibitors yielded the expected effects on YOD1 expressions, in agreement with the negative correlation observed between the expression levels of miR-221/222 and YOD1 in AML cells. Finally, overexpression of YOD1 stabilized p53, upregulated pro-apoptotic p53 downstream genes, and increased the sensitivity of AML cells to FLT3 inhibitors remarkably. Collectively, our study identified a pathway connecting miR-221/222, YOD1, and p53 in AML. Targeting miR-221/222 and stimulating YOD1 activity may improve the therapeutic effects of FLT3 inhibitors in patients with AML.
- Published
- 2023
- Full Text
- View/download PDF
3. Leukemogenic SHP2 mutations lead to erythropoietin independency of HCD-57 cells: a novel model for preclinical research of SHP2-mutant JMML
- Author
-
Yuming Zhao, Chunxiao He, Dengyang Zhang, Yao Guo, Zhiyong Peng, Liuting Yu, Na Li, Chun Chen, Zhizhuang Joe Zhao, and Yun Chen
- Subjects
SHP2 ,Cell model ,JMML ,HCD-57 ,Diseases of the blood and blood-forming organs ,RC633-647.5 ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Abstract Leukemogenic SHP2 mutations occur in 35% of patients with juvenile myelomonocytic leukemia (JMML), a rare but fatal hematopoietic malignancy without representative cell models, which are urgently needed to investigate the pathogenesis and to develop novel therapeutic strategies. In this study, we established stable cell lines with aberrant signaling resembling SHP2-mutant JMML through retroviral expression of SHP2-D61Y/E76K in HCD-57 cells, a murine erythroleukemia cell line that depends on erythropoietin (EPO) for survival. SHP2-D61Y/E76K drives the survival and proliferation of HCD-57 cells in the absence of EPO, but not in Ba/F3 cells in the absence of IL-3. Transformed HCD-57 cells showed activated MAPK signaling that is consistent with SHP2-mutant JMML. Transformed HCD-57 cells were sensitive to dasatinib and trametinib, two targeted drugs previously reported to inhibit SHP2-mutant JMML cells. Furthermore, we injected mutant SHP2-transformed HCD-57 cells into immune-deficient mice intravenously and found that these cells rapidly proliferated in the spleen and bone marrow, providing an excellent model for in vivo testing of drugs targeting the aberrant signaling of mutant SHP2. In conclusion, we established the novel cell lines HCD-57/SHP2-E76K and -D61Y that depended on signaling of mutant SHP2 for survival, thus resembling SHP2-mutant JMML. Our model is a valuable tool to investigate the pathogenic mechanisms of mutant SHP2 and targeted drugs for SHP2-mutant JMML.
- Published
- 2023
- Full Text
- View/download PDF
4. Bioinformatics analyses of combined databases identify shared differentially expressed genes in cancer and autoimmune disease
- Author
-
Yuan Sui, Shuping Li, Xue-Qi Fu, Zhizhuang Joe Zhao, and Shu Xing
- Subjects
Tumorigenesis ,Autoimmune genesis ,Invasive ductal carcinoma ,Systemic lupus erythematosus ,STAT1 ,OAS1 ,Medicine - Abstract
Abstract Background Inadequate immunity caused by poor immune surveillance leads to tumorigenesis, while excessive immunity due to breakdown of immune tolerance causes autoimmune genesis. Although the function of immunity during the onset of these two processes appears to be distinct, the underlying mechanism is shared. To date, gene expression data for large bodies of clinical samples are available, but the resemblances of tumorigenesis and autoimmune genesis in terms of immune responses remains to be summed up. Methods Considering the high disease prevalence, we chose invasive ductal carcinoma (IDC) and systemic lupus erythematosus (SLE) to study the potential commonalities of immune responses. We obtained gene expression data of IDC/SLE patients and normal controls from five IDC databases (GSE29044, GSE21422, GSE22840, GSE15852, and GSE9309) and five SLE databases (GSE154851, GSE99967, GSE61635, GSE50635, and GSE17755). We intended to identify genes differentially expressed in both IDC and SLE by using three bioinformatics tools including GEO2R, the limma R package, and Weighted Gene Co-expression Network Analysis (WGCNA) to perform function enrichment, protein-protein network, and signaling pathway analyses. Results The mRNA levels of signal transducer and activator of transcription 1 (STAT1), 2'-5'-oligoadenylate synthetase 1 (OAS1), 2'-5'-oligoadenylate synthetase like (OASL), and PML nuclear body scaffold (PML) were found to be differentially expressed in both IDC and SLE by using three different bioinformatics tools of GEO2R, the limma R package and WGCNA. From the combined databases in this study, the mRNA levels of STAT1 and OAS1 were increased in IDC while reduced in SLE. And the mRNA levels of OASL and PML were elevated in both IDC and SLE. Based on Kyoto Encyclopedia of Genes and Genomes pathway analysis and QIAGEN Ingenuity Pathway Analysis, both IDC and SLE were correlated with the changes of multiple components involved in the Interferon (IFN)-Janus kinase (JAK)-signal transducer and activator of transcription (STAT) signaling pathway. Conclusion The expression levels of STAT1 and OAS1 manifest the opposite expression tendency across cancer and autoimmune disease. They are components in the IFN-JAK-STAT signaling pathway related to both tumorigenesis and autoimmune genesis. STAT1 and OAS1-associated IFN-JAK-STAT signaling could explain the commonalities during tumorigenesis and autoimmune genesis and render significant information for more precise treatment from the point of immune homeostasis.
- Published
- 2023
- Full Text
- View/download PDF
5. Efficacy of SCF drug conjugate targeting c-KIT in gastrointestinal stromal tumor
- Author
-
Dengyang Zhang, Chunxiao He, Yao Guo, Jianfeng Li, Bo Li, Yuming Zhao, Liuting Yu, Zhiguang Chang, Hanzhong Pei, Ming Yang, Na Li, Qi Zhang, Yulong He, Yihang Pan, Zhizhuang Joe Zhao, Changhua Zhang, and Yun Chen
- Subjects
GIST ,SCF ,DM1 ,Targeted therapy ,Medicine - Abstract
Abstract Background Gastrointestinal stromal tumor (GIST) is a rare type of cancer that occurs in the gastrointestinal tract. The majority of GIST cases carry oncogenic forms of KIT, the receptor for stem cell factor (SCF). Small molecule kinase inhibitor imatinib is effective in prolonging the survival of GIST patients by targeting KIT. However, drug resistance often develops during the therapeutic treatment. Here, we produced a SCF-emtansine drug conjugate (SCF-DM1) with favorable drug efficacy towards GIST cells. Methods Recombinant human SCF (rhSCF) was expressed in E. coli cells and further purified with Ni–NTA Sepharose and Phenyl Sepharose. It was then conjugated with DM1, and the conjugated product SCF-DM1 was evaluated using in vitro cell-based assays and in vivo xenograft mouse model. Results SCF-DM1 was effective in inhibiting imatinib-sensitive and -resistant GIST cell lines and primary tumor cells, with IC50 values of
- Published
- 2022
- Full Text
- View/download PDF
6. Editorial: Linking cellular metabolism to hematological malignancies
- Author
-
Xiujing He, Manoj Kumar Kashyap, Zhizhuang Joe Zhao, Jian Yu, and Hubing Shi
- Subjects
hematological malignancies ,cellular metabolism ,metabolic reprogramming ,immunometabolism ,drug resistance ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Published
- 2022
- Full Text
- View/download PDF
7. Expression of a recombinant FLT3 ligand and its emtansine conjugate as a therapeutic candidate against acute myeloid leukemia cells with FLT3 expression
- Author
-
Dengyang Zhang, Yao Guo, Yuming Zhao, Liuting Yu, Zhiguang Chang, Hanzhong Pei, Junbin Huang, Chun Chen, Hongman Xue, Xiaojun Xu, Yihang Pan, Ningning Li, Chengming Zhu, Zhizhuang Joe Zhao, Jian Yu, and Yun Chen
- Subjects
FLT3 ,FL ,AML ,DM1 ,Targeted therapy ,Microbiology ,QR1-502 - Abstract
Abstract Background Most patients with acute myeloid leukemia (AML) remain uncurable and require novel therapeutic methods. Gain-of-function FMS-like tyrosine kinase 3 (FLT3) mutations are present in 30–40% of AML patients and serve as an attractive therapeutic target. In addition, FLT3 is aberrantly expressed on blasts in > 90% of patients with AML, making the FLT3 ligand-based drug conjugate a promising therapeutic strategy for the treatment of patients with AML. Here, E. coli was used as a host to express recombinant human FLT3 ligand (rhFL), which was used as a specific vehicle to deliver cytotoxic drugs to FLT3 + AML cells. Methods Recombinant hFL was expressed and purified from induced recombinant BL21 (DE3) E. coli. Purified rhFL and emtansine (DM1) were conjugated by an N-succinimidyl 3-(2-pyridyldithio)propionate (SPDP) linker. We evaluated the potency of the conjugation product FL-DM1 against FLT3-expressing AML cells by examining viability, apoptosis and the cell cycle. The activation of proteins related to the activation of FLT3 signaling and apoptosis pathways was detected by immunoblotting. The selectivity of FL-DM1 was assessed in our unique HCD-57 cell line, which was transformed with the FLT3 internal tandem duplication mutant (FLT3-ITD). Results Soluble rhFL was successfully expressed in the periplasm of recombinant E. coli. The purified rhFL was bioactive in stimulating FLT3 signaling in AML cells, and the drug conjugate FL-DM1 showed activity in cell signaling and internalization. FL-DM1 was effective in inhibiting the survival of FLT3-expressing THP-1 and MV-4-11 AML cells, with half maximal inhibitory concentration (IC50) of 12.9 nM and 1.1 nM. Additionally, FL-DM1 induced caspase-3-dependent apoptosis and arrested the cell cycle at the G2/M phase. Moreover, FL-DM1 selectively targeted HCD-57 cells transformed by FLT3-ITD but not parental HCD-57 cells without FLT3 expression. FL-DM1 can also induce obvious apoptosis in primary FLT3-positive AML cells ex vivo. Conclusions Our data demonstrated that soluble rhFL can be produced in a bioactive form in the periplasm of recombinant E. coli. FL can be used as a specific vehicle to deliver DM1 into FLT3-expressing AML cells. FL-DM1 exhibited cytotoxicity in FLT3-expressing AML cell lines and primary AML cells. FL-DM1 may have potential clinical applications in treating patients with FLT3-positive AML.
- Published
- 2021
- Full Text
- View/download PDF
8. Development of a highly sensitive method for detection of FLT3D835Y
- Author
-
Yao Guo, Honghua Sun, Dengyang Zhang, Yuming Zhao, Mingxia Shi, Ming Yang, Shu Xing, Xueqi Fu, Ting Bin, Bo Lu, Shunjie Wu, Xiaojun Xu, Xuesong Xu, Yun Chen, and Zhizhuang Joe Zhao
- Subjects
Tyrosine kinase ,Acute myeloid leukemia ,FLT3-TKD ,Detection of mutations ,Therapeutics. Pharmacology ,RM1-950 - Abstract
Abstract Background Acute myeloid leukemia (AML) is a malignant hematological neoplasm of myeloid progenitor cells. Mutations of FLT3 in its tyrosine kinase domain (FLT3-TKD) are found in ~ 8% of patients with AML, with D835Y as the most common substitution. This mutation activates survival signals that drives the disease and is resistant to the first generation FLT3 inhibitors. Development of a highly sensitive method to detect FLT3D835Y is important to direct therapeutic options, predict prognosis, and monitor minimal residual disease in patients with AML. Methods and results In the present study, we developed a highly sensitive FLT3D835Y detection method by using the restriction fragment nested allele-specific PCR technique. The method consists of three steps: 1) initial amplification of DNA samples with PCR primers surrounding the FLT3D835Y mutation site, 2) digestion of the PCR products with restriction enzyme EcoRV that only cleaves the wild type allele, and 3) detection of FLT3D835Y by allele-specific PCR with nested primers. We were able to detect FLT3D835Y with a sensitivity of 0.001% by using purified plasmid DNAs and blood cell DNAs containing known proportions of FLT3D835Y. We analyzed blood cell DNA samples from 64 patients with AML and found six FLT3D835Y-positive cases, two of which could not be detected by conventional DNA sequencing methods. Importantly, the method was able to detect FLT3D835Y in a sample collected from a relapsed patient while the patient was in complete remission with negative MRD determined by flow cytometry. Therefore, our RFN-AS-PCR detected MRD after treatment that was missed by flow cytometry and Sanger DNA sequencing, by conventional methods. Conclusions We have developed a simple and highly sensitive method that will allow for detection of FLT3D835Y at a very low level. This method may have major clinical implications for treatment of AML.
- Published
- 2020
- Full Text
- View/download PDF
9. Bidirectional Interaction Between Cancer Cells and Platelets Provides Potential Strategies for Cancer Therapies
- Author
-
Liuting Yu, Yao Guo, Zhiguang Chang, Dengyang Zhang, Shiqiang Zhang, Hanzhong Pei, Jun Pang, Zhizhuang Joe Zhao, and Yun Chen
- Subjects
platelets ,thrombosis ,cancer cells ,cancer-platelet crosstalk ,cancer treatment ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Platelets are essential components in the tumor microenvironment. For decades, clinical data have demonstrated that cancer patients have a high risk of thrombosis that is associated with adverse prognosis and decreased survival, indicating the involvement of platelets in cancer progression. Increasing evidence confirms that cancer cells are able to induce production and activation of platelets. Once activated, platelets serve as allies of cancer cells in tumor growth and metastasis. They can protect circulating tumor cells (CTCs) against the immune system and detachment-induced apoptosis while facilitating angiogenesis and tumor cell adhesion and invasion. Therefore, antiplatelet agents and platelet-based therapies should be developed for cancer treatment. Here, we discuss the mechanisms underlying the bidirectional cancer-platelet crosstalk and platelet-based therapeutic approaches.
- Published
- 2021
- Full Text
- View/download PDF
10. Tyrosine Kinase ROR1 as a Target for Anti-Cancer Therapies
- Author
-
Yuming Zhao, Dengyang Zhang, Yao Guo, Bo Lu, Zhizhuang Joe Zhao, Xiaojun Xu, and Yun Chen
- Subjects
ROR1 ,kinase activity ,kinase function ,small molecule inhibitors ,targeted therapy ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
Receptor tyrosine kinase ROR1 plays an essential role in embryogenesis and is overexpressed in many types of malignant tumors. Studies have demonstrated that it plays an important role in oncogenesis by activating cell survival signaling events, particularly the non-canonical WNT signaling pathway. Antibody-based immunotherapies targeting ROR1 have been developed and evaluated in preclinical and clinical studies with promising outcomes. However, small molecule inhibitors targeting ROR1 are underappreciated because of the initial characterization of ROR1 as a peusdokinase. The function of ROR1 as a tyrosine kinase remains poorly understood, although accumulating evidence have demonstrated its intrinsic tyrosine kinase activity. In this review, we analyzed the structural and functional features of ROR1 and discussed therapeutic strategies targeting this kinase.
- Published
- 2021
- Full Text
- View/download PDF
11. Association of PTPN22-C1858T Polymorphism With Susceptibility to Mycobacterium tuberculosis and Mycobacterium leprae Infection: A Meta-Analysis
- Author
-
Shuping Li, Xiaohua Wang, Yuming Zhao, Juan Yang, Tianjiao Cui, Zhizhuang Joe Zhao, Yun Chen, and Zhihua Zheng
- Subjects
PTPN22-C1858T ,single-nucleotide polymorphism ,tuberculosis ,leprosy ,Mycobacterium tuberculosis ,Mycobacterium leprae ,Immunologic diseases. Allergy ,RC581-607 - Abstract
It was previously published that single-nucleotide polymorphism rs2476601 (PTPN22 [protein tyrosine phosphatase non-receptor type 22]-C1858T) might be related to increased sensibility to Mycobacterium tuberculosis and M. leprae infection. However, the results were inconclusive despite a high degree of similarity between both parameters. Herein, we carried out this meta-analysis to systematically summarize and articulate the correlation between PTPN22-C1858T polymorphism and mycobacterial infection. The susceptibility of PTPN22-C1858T carriers with autoimmune conditions receiving immunosuppressive therapy to M. tuberculosis and M. leprae infection was determined. A systematic retrieval of studies on relevance of PTPN22-C1858T polymorphism to susceptibility of M. tuberculosis or M. leprae infection was performed in Chinese National Knowledge Infrastructure, PubMed and Embase databases. We regarded Odds ratios (ORs) and 95% confidence intervals (CIs) as the determined effect size. Finally, four and two case-control studies on tuberculosis and leprosy, respectively, were included. In all genetic models, without indicated association between PTPN22-C1858T polymorphism and tuberculosis’s susceptibility. [C versus T: OR = 0.22 (95% CI: 0.09–0.50, PH = 0.887); CT versus CC: OR = 0.21 (95% CI: 0.09–0.49, PH = 0.889); TT+CT versus CC: OR = 0.21 (95% CI: 0.09–0.49, PH = 0.889)]. A significantly increased risk of leprosy was perceived in patients with the PTPN22-C1858T polymorphism [C versus T: OR = 2.82 (95% CI: 1.02–7.81, PH = 0.108)]. While the PTPN22-C1858T polymorphism is irrelevant to higher susceptibility to the infection of M. tuberculosis in Caucasians and Asians, it is relevant to increased susceptibility to the infection of M. leprae. However, the results of M. leprae are supposed to interpreted with prudence owing to the limited quantity of studies and heterogeneity. Further well-designed studies with sufficient populations are required to verify our conclusions.
- Published
- 2021
- Full Text
- View/download PDF
12. The Jak2 Inhibitor, G6, Alleviates Jak2-V617F–Mediated Myeloproliferative Neoplasia by Providing Significant Therapeutic Efficacy to the Bone Marrow
- Author
-
Annet Kirabo, Sung O. Park, Anurima Majumder, Meghanath Gali, Mary K. Reinhard, Heather L. Wamsley, Zhizhuang Joe Zhao, Christopher R. Cogle, Kirpal S. Bisht, György M. Keserű, and Peter P. Sayeski
- Subjects
Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,RC254-282 - Abstract
We recently developed a Janus kinase 2 (Jak2) small-molecule inhibitor called G6 and found that it inhibits Jak2-V617F– mediated pathologic cell growth in vitro, ex vivo, and in vivo. However, its ability to inhibit Jak2-V617F–mediated myeloproliferative neoplasia, with particular emphasis in the bone marrow, has not previously been examined. Here, we investigated the efficacy of G6 in a transgenic mouse model of Jak2-V617F–mediated myeloproliferative neoplasia. We found that G6 provided therapeutic benefit to the peripheral blood as determined by elimination of leukocytosis, thrombocytosis, and erythrocytosis. G6 normalized the pathologically high plasma concentrations of interleukin 6 (IL-6). In the liver, G6 eliminated Jak2-V617F–driven extramedullary hematopoiesis. With respect to the spleen, G6 significantly reduced both the spleno-megaly and megakaryocytic hyperplasia. In the critically important bone marrow, G6 normalized the pathologically high levels of phospho-Jak2 and phospho–signal transducer and activator of transcription 5 (STAT5). It significantly reduced the megakaryocytic hyperplasia in the marrow and completely normalized the M/E ratio. Most importantly, G6 selectively reduced the mutant Jak2 burden by 67% on average, with virtual elimination of mutant Jak2 cells in one third of all treated mice. Lastly, clonogenic assays using marrow stem cells from the myeloproliferative neoplasm mice revealed a time-dependent elimination of the clonogenic growth potential of these cells by G6. Collectively, these data indicate that G6 exhibits exceptional efficacy in the peripheral blood, liver, spleen, and, most importantly, in the bone marrow, thereby raising the possibility that this compound may alter the natural history of Jak2-V617F–mediated myeloproliferative neoplasia.
- Published
- 2011
- Full Text
- View/download PDF
13. Generation and characterization of a JAK2V617F-containing erythroleukemia cell line.
- Author
-
Wanke Zhao, Kang Zou, Taleah Farasyn, Wanting Tina Ho, and Zhizhuang Joe Zhao
- Subjects
Medicine ,Science - Abstract
The JAK2V617F mutation is found in the majority of patients with myeloproliferative neoplasms (MPNs). Transgenic expression of the mutant gene causes MPN-like phenotypes in mice. We have produced JAK2V617F mice with p53 null background. Some of these mice developed acute erythroleukemia. From one of these mice, we derived a cell line designated J53Z1. J53Z1 cells were stained positive for surface markers CD71 and CD117 but negative for Sca-1, TER-119, CD11b, Gr-1, F4/80, CD11c, CD317, CD4, CD8a, CD3e, B220, CD19, CD41, CD42d, NK-1.1, and FceR1. Real time PCR analyses demonstrated expressions of erythropoietin receptor EpoR, GATA1, and GATA2 in these cells. J53Z1 cells grew rapidly in suspension culture containing fetal bovine serum with a doubling time of ∼18 hours. When transplanted into C57Bl/6 mice, J53Z1 cells induced acute erythroleukemia with massive infiltration of tumor cells in the spleen and liver. J53Z1 cells were responsive to stimulation with erythropoietin and stem cell factor and were selectively inhibited by JAK2 inhibitors which induced apoptosis of the cells. Together, J53Z1 cells belong to the erythroid lineage, and they may be useful for studying the role of JAK2V617F in proliferation and differentiation of erythroid cells and for identifying potential therapeutic drugs targeting JAK2.
- Published
- 2014
- Full Text
- View/download PDF
14. Stem and progenitor cell subsets are affected by JAK2 signaling and can be monitored by flow cytometry.
- Author
-
Ryuji Iida, Robert S Welner, Wanke Zhao, José Alberola-lla, Kay L Medina, Zhizhuang Joe Zhao, and Paul W Kincade
- Subjects
Medicine ,Science - Abstract
Although extremely rare, hematopoietic stem cells (HSCs) are divisible into subsets that differ with respect to differentiation potential and cell surface marker expression. For example, we recently found that CD86(-) CD150(+) CD48(-) HSCs have limited potential for lymphocyte production. This could be an important new tool for studying hematological abnormalities. Here, we analyzed HSC subsets with a series of stem cell markers in JAK2V617F transgenic (Tg) mice, where the mutation is sufficient to cause myeloproliferative neoplasia with lymphocyte deficiency. Total numbers of HSC were elevated 3 to 20 fold in bone marrow of JAK2V617F mice. Careful analysis suggested the accumulation involved multiple HSC subsets, but particularly those characterized as CD150(HI) CD86(-) CD18(L)°CD41(+) and excluding Hoechst dye. Real-Time PCR analysis of their HSC revealed that the erythropoiesis associated gene transcripts Gata1, Klf1 and Epor were particularly high. Flow cytometry analyses based on two differentiation schemes for multipotent progenitors (MPP) also suggested alteration by JAK2 signals. The low CD86 on HSC and multipotent progenitors paralleled the large reductions we found in lymphoid progenitors, but the few that were produced functioned normally when sorted and placed in culture. Either of two HSC subsets conferred disease when transplanted. Thus, flow cytometry can be used to observe the influence of abnormal JAK2 signaling on stem and progenitor subsets. Markers that similarly distinguish categories of human HSCs might be very valuable for monitoring such conditions. They could also serve as indicators of HSC fitness and suitability for transplantation.
- Published
- 2014
- Full Text
- View/download PDF
15. Aconitase regulation of erythropoiesis correlates with a novel licensing function in erythropoietin-induced ERK signaling.
- Author
-
Anne-Laure Talbot, Grant C Bullock, Lorrie L Delehanty, Martin Sattler, Zhizhuang Joe Zhao, and Adam N Goldfarb
- Subjects
Medicine ,Science - Abstract
Erythroid development requires the action of erythropoietin (EPO) on committed progenitors to match red cell output to demand. In this process, iron acts as a critical cofactor, with iron deficiency blunting EPO-responsiveness of erythroid progenitors. Aconitase enzymes have recently been identified as possible signal integration elements that couple erythropoiesis with iron availability. In the current study, a regulatory role for aconitase during erythropoiesis was ascertained using a direct inhibitory strategy.In C57BL/6 mice, infusion of an aconitase active-site inhibitor caused a hypoplastic anemia and suppressed responsiveness to hemolytic challenge. In a murine model of polycythemia vera, aconitase inhibition rapidly normalized red cell counts, but did not perturb other lineages. In primary erythroid progenitor cultures, aconitase inhibition impaired proliferation and maturation but had no effect on viability or ATP levels. This inhibition correlated with a blockade in EPO signal transmission specifically via ERK, with preservation of JAK2-STAT5 and Akt activation. Correspondingly, a physical interaction between ERK and mitochondrial aconitase was identified and found to be sensitive to aconitase inhibition.Direct aconitase inhibition interferes with erythropoiesis in vivo and in vitro, confirming a lineage-selective regulatory role involving its enzymatic activity. This inhibition spares metabolic function but impedes EPO-induced ERK signaling and disturbs a newly identified ERK-aconitase physical interaction. We propose a model in which aconitase functions as a licensing factor in ERK-dependent proliferation and differentiation, thereby providing a regulatory input for iron in EPO-dependent erythropoiesis. Directly targeting aconitase may provide an alternative to phlebotomy in the treatment of polycythemia vera.
- Published
- 2011
- Full Text
- View/download PDF
16. SHP2 participates in decidualization by activating ERK to maintain normal nuclear localization of progesterone receptor
- Author
-
Lin Chen, Weijie Zhao, Mengxiong Li, Yazhu Yang, Chengzi Tian, Dengyang Zhang, Zhiguang Chang, Yunzhe Zhang, Zhizhuang Joe Zhao, Yun Chen, and Lin Ma
- Subjects
Embryology ,Endocrinology ,Reproductive Medicine ,Obstetrics and Gynecology ,Cell Biology - Abstract
Decidualization is the process of conversion of endometrial stromal cells (ESCs) into decidual stromal cells (DSCs), which is caused by progesterone production that begins during the luteal phase of the menstrual cycle and then increases throughout pregnancy dedicated to support embryonic development. Decidualization deficiency is closely associated with various pregnancy complications, such as recurrent miscarriage (RM). Here, we reported that Src-homology-2-containing phospho-tyrosine phosphatase (SHP2), a key regulator in the signal transduction process downstream of various receptors, plays an indispensable role in decidualization. SHP2 expression was upregulated during decidualization. SHP2 inhibitor RMC-4550 and shRNA mediated SHP2 reduction resulted in a decreased level of phosphorylation of ERK and aberrant cytoplasmic localization of progesterone receptor (PR), coinciding with reduced expression of IGFBP1 and various other target genes of decidualization. Solely inhibiting ERK activity recapitulated these observations. Administration of RMC-4550 led to decidualization deficiency and embryo absorption in mouse. Moreover, reduced expression of SHP2 was detected in decidua of RM patients. Our results revealed that SHP2 is key to PR's nuclear localization, thereby indispensable for decidualization and that reduced expression of SHP2 might be engaged in the pathogenesis of RM.
- Published
- 2023
- Full Text
- View/download PDF
17. Sunitinib selectively targets leukemogenic signaling of mutant SHP2 in juvenile myelomonocytic leukemia
- Author
-
Chunxiao He, Zhiyong Peng, Dengyang Zhang, Yao Guo, Tianqi Liang, Yuming Zhao, Liuting Yu, Qi Zhang, Zhiguang Chang, Yan Xiao, Na Li, Hongman Xue, Shunjie Wu, Zhizhuang Joe Zhao, Changhua Zhang, and Yun Chen
- Subjects
Pharmacology ,Biochemistry - Published
- 2023
- Full Text
- View/download PDF
18. Bidirectional Interaction Between Cancer Cells and Platelets Provides Potential Strategies for Cancer Therapies
- Author
-
Hanzhong Pei, Zhiguang Chang, Yun Chen, Liuting Yu, Shiqiang Zhang, Zhizhuang Joe Zhao, Dengyang Zhang, Yao Guo, and Jun Pang
- Subjects
Cancer Research ,Tumor microenvironment ,Angiogenesis ,business.industry ,Cancer ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Review ,cancer-platelet crosstalk ,medicine.disease ,cancer treatment ,Metastasis ,Circulating tumor cell ,Immune system ,Oncology ,Cancer cell ,platelets ,medicine ,Cancer research ,cancer cells ,Platelet ,business ,thrombosis ,RC254-282 - Abstract
Platelets are essential components in the tumor microenvironment. For decades, clinical data have demonstrated that cancer patients have a high risk of thrombosis that is associated with adverse prognosis and decreased survival, indicating the involvement of platelets in cancer progression. Increasing evidence confirms that cancer cells are able to induce production and activation of platelets. Once activated, platelets serve as allies of cancer cells in tumor growth and metastasis. They can protect circulating tumor cells (CTCs) against the immune system and detachment-induced apoptosis while facilitating angiogenesis and tumor cell adhesion and invasion. Therefore, antiplatelet agents and platelet-based therapies should be developed for cancer treatment. Here, we discuss the mechanisms underlying the bidirectional cancer-platelet crosstalk and platelet-based therapeutic approaches.
- Published
- 2021
- Full Text
- View/download PDF
19. Aurora Kinase B/C Inhibitor GSK1070916 Specifically Targets Juvenile Myelomonocytic Leukemia Cells with SHP2(PTPN11) Mutation
- Author
-
Chunxiao He, Dengyang Zhang, Yao Guo, Yuming Zhao, Junbin Huang, Hongman Xue, Chun Chen, Yun Chen, and Zhizhuang Joe Zhao
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
20. The Tubular Cells Produce Fms-like Tyrosine Kinase Receptor 3 Ligand to Recover Kidney Function after Acute Kidney Injury Disease
- Author
-
Na Li, Yao Guo, Zhihua Zheng, Yun Chen, and Zhizhuang Joe Zhao
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
21. Niclosamide Inhibits the Growth of Acute Lymphoblastic Leukemia Cells By Increasing Intracellular ROS and Activation of p53-PUMA/NOXA
- Author
-
Junbin Huang, Jing Yang, Yun Chen, Zhizhuang Joe Zhao, and Chun Chen
- Subjects
Immunology ,Cell Biology ,Hematology ,Biochemistry - Published
- 2022
- Full Text
- View/download PDF
22. HMGA1 chromatin regulators induce transcriptional networks involved in GATA2 and proliferation during MPN progression
- Author
-
Liping Li, Jung-Hyun Kim, Wenyan Lu, Donna M. Williams, Joseph Kim, Leslie Cope, Raajit K. Rampal, Richard P. Koche, Lingling Xian, Li Z. Luo, Marija Vasiljevic, Daniel R. Matson, Zhizhuang Joe Zhao, Ophelia Rogers, Matthew C. Stubbs, Karen Reddy, Antonio-Rodriguez Romero, Bethan Psaila, Jerry L. Spivak, Alison R. Moliterno, and Linda M. S. Resar
- Subjects
Myeloid Neoplasia ,Myeloproliferative Disorders ,Immunology ,Cell Biology ,Hematology ,Janus Kinase 2 ,Biochemistry ,Chromatin ,GATA2 Transcription Factor ,Leukemia, Myeloid, Acute ,Mice ,Primary Myelofibrosis ,Humans ,Animals ,Gene Regulatory Networks ,HMGA1a Protein ,Transcription Factors ,Cell Proliferation - Abstract
Myeloproliferative neoplasms (MPNs) transform to myelofibrosis (MF) and highly lethal acute myeloid leukemia (AML), although the actionable mechanisms driving progression remain elusive. Here, we elucidate the role of the high mobility group A1 (HMGA1) chromatin regulator as a novel driver of MPN progression. HMGA1 is upregulated in MPN, with highest levels after transformation to MF or AML. To define HMGA1 function, we disrupted gene expression via CRISPR/Cas9, short hairpin RNA, or genetic deletion in MPN models. HMGA1 depletion in JAK2V617F AML cell lines disrupts proliferation, clonogenicity, and leukemic engraftment. Surprisingly, loss of just a single Hmga1 allele prevents progression to MF in JAK2V617F mice, decreasing erythrocytosis, thrombocytosis, megakaryocyte hyperplasia, and expansion of stem and progenitors, while preventing splenomegaly and fibrosis within the spleen and BM. RNA-sequencing and chromatin immunoprecipitation sequencing revealed HMGA1 transcriptional networks and chromatin occupancy at genes that govern proliferation (E2F, G2M, mitotic spindle) and cell fate, including the GATA2 master regulatory gene. Silencing GATA2 recapitulates most phenotypes observed with HMGA1 depletion, whereas GATA2 re-expression partially rescues leukemogenesis. HMGA1 transactivates GATA2 through sequences near the developmental enhancer (+9.5), increasing chromatin accessibility and recruiting active histone marks. Further, HMGA1 transcriptional networks, including proliferation pathways and GATA2, are activated in human MF and MPN leukemic transformation. Importantly, HMGA1 depletion enhances responses to the JAK2 inhibitor, ruxolitinib, preventing MF and prolonging survival in murine models of JAK2V617F AML. These findings illuminate HMGA1 as a key epigenetic switch involved in MPN transformation and a promising therapeutic target to treat or prevent disease progression.
- Published
- 2021
23. Loss of tyrosine phosphatase SHP2 activity promotes growth of colorectal carcinoma HCT-116 cells
- Author
-
Zhizhuang Joe Zhao, Xin Chen, Wanting Tina Ho, Xueqi Fu, Wanke Zhao, and Shu Xing
- Subjects
Cell biology ,Cancer Research ,Letter ,Drug discovery ,Carcinogenesis ,Colorectal cancer ,Chemistry ,lcsh:R ,lcsh:Medicine ,Protein Tyrosine Phosphatase, Non-Receptor Type 11 ,HCT116 Cells ,medicine.disease ,Gene Expression Regulation, Neoplastic ,lcsh:Biology (General) ,TYROSINE PHOSPHATASE SHP2 ,Genetics ,Cancer research ,medicine ,Humans ,Colorectal Neoplasms ,lcsh:QH301-705.5 ,Cell Proliferation - Published
- 2020
- Full Text
- View/download PDF
24. ASXL1 alteration cooperates with JAK2V617F to accelerate myelofibrosis
- Author
-
Peng Zhang, Zhizhuang Joe Zhao, Jie Bai, Yuan Zhou, Mingjiang Xu, Shi Chen, Stephen D. Nimer, Feng Chun Yang, Hui Yang, Shohei Yamatomo, and Ying Guo
- Subjects
Regulation of gene expression ,Cancer Research ,Extramural ,business.industry ,Disease progression ,Amino acid substitution ,Hematology ,medicine.disease ,Oncology ,Mutation (genetic algorithm) ,Cancer research ,medicine ,Epistasis ,Myelofibrosis ,business - Published
- 2019
- Full Text
- View/download PDF
25. Tyrosine Kinase ROR1 as a Target for Anti-Cancer Therapies
- Author
-
Yao Guo, Dengyang Zhang, Xiaojun Xu, Yun Chen, Zhizhuang Joe Zhao, Yuming Zhao, and Bo Lu
- Subjects
Cancer Research ,kinase function ,biology ,Kinase ,medicine.medical_treatment ,Wnt signaling pathway ,Neoplasms. Tumors. Oncology. Including cancer and carcinogens ,Review ,kinase activity ,medicine.disease_cause ,targeted therapy ,small molecule inhibitors ,Receptor tyrosine kinase ,Targeted therapy ,Oncology ,ROR1 ,biology.protein ,medicine ,Cancer research ,Kinase activity ,Carcinogenesis ,Tyrosine kinase ,RC254-282 - Abstract
Receptor tyrosine kinase ROR1 plays an essential role in embryogenesis and is overexpressed in many types of malignant tumors. Studies have demonstrated that it plays an important role in oncogenesis by activating cell survival signaling events, particularly the non-canonical WNT signaling pathway. Antibody-based immunotherapies targeting ROR1 have been developed and evaluated in preclinical and clinical studies with promising outcomes. However, small molecule inhibitors targeting ROR1 are underappreciated because of the initial characterization of ROR1 as a peusdokinase. The function of ROR1 as a tyrosine kinase remains poorly understood, although accumulating evidence have demonstrated its intrinsic tyrosine kinase activity. In this review, we analyzed the structural and functional features of ROR1 and discussed therapeutic strategies targeting this kinase.
- Published
- 2021
- Full Text
- View/download PDF
26. Additional file 1 of Expression of a recombinant FLT3 ligand and its emtansine conjugate as a therapeutic candidate against acute myeloid leukemia cells with FLT3 expression
- Author
-
Dengyang Zhang, Guo, Yao, Yuming Zhao, Liuting Yu, Zhiguang Chang, Hanzhong Pei, Junbin Huang, Chen, Chun, Hongman Xue, Xiaojun Xu, Yihang Pan, Ningning Li, Chengming Zhu, Zhizhuang Joe Zhao, Yu, Jian, and Chen, Yun
- Abstract
Additional file 1. LC-MS materials, methods and results: S1. Chemicals and Instrumentation; S2. Sample Preparation; S3: LC-MS Analysis method; S4: LC-MS analysis results
- Published
- 2021
- Full Text
- View/download PDF
27. FLT3-TKD in the Prognosis of Patients With Acute Myeloid Leukemia: A Meta-Analysis
- Author
-
Yun Chen, Chun Chen, Jinqiu Yuan, Yiqing Zhang, Ming Yang, Bo Lu, Chenju Yi, Xiaohua Wang, Xiaojun Xu, Zhizhuang Joe Zhao, Shuping Li, Yuming Zhao, Zhihua Zheng, Yan Lei, and Dongjun Lin
- Subjects
Oncology ,medicine.medical_specialty ,business.industry ,Daunorubicin ,Hazard ratio ,Subgroup analysis ,Publication bias ,Gene mutation ,hemic and lymphatic diseases ,Meta-analysis ,Internal medicine ,embryonic structures ,medicine ,Cytarabine ,Prospective cohort study ,business ,medicine.drug - Abstract
Background: Fms-like tyrosine kinase 3 (FLT3) gene mutations occur in approximately 30% of all AML patients. Internal tandem duplication (ITD) in the juxtamembrane domain and point mutations within the tyrosine kinase domain (TKD) are two distinct types of FLT3 mutations. FLT3-ITD has been determined as an independent poor prognostic factor, but the prognostic impact of FLT3-TKD remains controversial. Hence, we performed a meta-analysis to investigate the prognostic significance of FLT3-TKD in patients with AML. Methods: A systematic retrieval of studies on FLT3-TKD in patients with AML was performed in PubMed, Embase, and CNKI databases on Sept. 30th, 2020. Hazard ratio (HR) and its 95% confidence intervals (95% CIs) were used to determine the effect size. Meta-regression model and subgroup analysis were used for heterogeneity analysis. Begg’s and Egger’s tests were performed to detect potential publication bias. The sensitivity analysis was performed to evaluate the stability of findings in meta-analysis. Findings: Twenty prospective cohort studies (n=10970) on the prognostic effect of FLT3-TKD in AML were included: 9744 subjects with FLT3-WT and 1226 subjects with FLT3-TKD. We found that FLT3-TKD revealed no significant effect on DFS (HR=1.12, 95% CI: 0.90-1.41) and OS (HR=0.98, 95% CI: 0.76-1.27) in general. However, meta-regressions demonstrated that ethnicity contributed to the high heterogeneity observed in the prognosis of FLT3-TKD in AML. To be specific, FLT3-TKD represented a beneficial prognosis of DFS (HR=0.56, 95% CI: 0.37-0.85) and OS (HR=0.63, 95% CI: 0.42-0.95) for Asians while it represented an adverse prognosis of DFS for Caucasians with AML (HR=1.34, 95% CI: 1.07-1.67). Interpretation: Our results showed that FLT3-TKD revealed no significant effects on DFS and OS of patients with AML, which is consistent with the controversial status nowadays. Distinct prognosis of FLT3-TKD in AML between Asians and Caucasians may attribute to different chemotherapy regimens (including the dose of cytarabine and daunorubicin in conventional "3+7" induction and the clinical usage of HHT) and diverse genetic background. The dose of cytarabine (100 mg/m2) and daunorubicin (50 mg/m2) used in Asian countries in "3+7" induction therapy may benefit Caucasians with AML carrying FLT3-TKD. Funding Information: National Natural Science Foundation of China (NSFC, Grant No. 82000150). Declaration of Interests: We declare no competing interests.
- Published
- 2021
- Full Text
- View/download PDF
28. Enhanced RIPK3 Kinase Activity-dependent Lytic Cell Death in M1 but Not M2 Macrophages
- Author
-
Zhizhuang Joe Zhao, Steven Idell, Hua Tang, Suman Kundu, Qin Hao, and Joshua Kleam
- Subjects
0301 basic medicine ,Programmed cell death ,Necroptosis ,Immunology ,Inflammation ,Article ,03 medical and health sciences ,Mice ,0302 clinical medicine ,medicine ,Macrophage ,Animals ,DNA, Z-Form ,Benzothiazoles ,RNA, Messenger ,Kinase activity ,Phosphorylation ,Molecular Biology ,Cells, Cultured ,Innate immune system ,Cell Death ,L-Lactate Dehydrogenase ,Effector ,Kinase ,Chemistry ,Macrophages ,RNA-Binding Proteins ,Macrophage Activation ,Cell biology ,Up-Regulation ,Mice, Inbred C57BL ,030104 developmental biology ,RAW 264.7 Cells ,Receptor-Interacting Protein Serine-Threonine Kinases ,Quinolines ,medicine.symptom ,Protein Kinases ,030215 immunology ,Signal Transduction - Abstract
Macrophages play a crucial role in host innate immune defense against infection and tissue injury. Macrophages are highly plastic cells and their subtypes have been characterized as M1 (also termed classically activated) and M2 (alternatively activated). Although the M1/M2 paradigm has been well documented, less is known regarding the role of macrophage activation/polarization in inflammation-associated necrotic cell death. To address this gap in current knowledge, we prepared bone marrow-derived macrophages, induced them to M1 or M2 subtypes, and then investigated the expression of necroptosis signaling molecules and macrophage subtype-dependent responses to different necroptosis inducers. We found that necroptosis effector mixed lineage kinase domain-like protein (MLKL) and the key necroptosis regulator Z-DNA/RNA binding protein 1 were predominantly induced in M1 but not M2 macrophages. Interestingly, the protein but not mRNA levels of receptor-interacting protein kinase-3 (RIPK3) were also upregulated in M1 macrophages. We further found that macrophage necrotic cell death, the releases of lactate dehydrogenase and dead cell proteases as well as MLKL phosphorylation at Ser345 in response to various necroptosis inducers were greatly augmented in M1 but not M2 macrophages, and the accelerated effects were blocked by two structurally distinct specific RIPK3 inhibitors GSK872 or GSK843. Thus, our findings demonstrate that M1 but not M2 subtypes of macrophages are more susceptible to inflammation-related lytic cell death in an RIPK3 kinase activity-dependent manner.
- Published
- 2020
29. Development of a highly sensitive method for detection of FLT3D835Y
- Author
-
Dengyang Zhang, Xuesong Xu, Yun Chen, Shunjie Wu, Shu Xing, Zhizhuang Joe Zhao, Yuming Zhao, Bo Lu, Xiaojun Xu, Ming Yang, Xueqi Fu, Honghua Sun, Ting Bin, Mingxia Shi, and Yao Guo
- Subjects
0301 basic medicine ,Clinical Biochemistry ,medicine.disease_cause ,DNA sequencing ,Restriction fragment ,03 medical and health sciences ,symbols.namesake ,Detection of mutations ,0302 clinical medicine ,Plasmid ,hemic and lymphatic diseases ,FLT3-TKD ,medicine ,Tyrosine kinase ,Sanger sequencing ,Mutation ,Acute myeloid leukemia ,biology ,lcsh:RM1-950 ,Biochemistry (medical) ,Methodology ,Myeloid leukemia ,Molecular biology ,Minimal residual disease ,Restriction enzyme ,lcsh:Therapeutics. Pharmacology ,030104 developmental biology ,030220 oncology & carcinogenesis ,biology.protein ,symbols ,Molecular Medicine - Abstract
Background Acute myeloid leukemia (AML) is a malignant hematological neoplasm of myeloid progenitor cells. Mutations of FLT3 in its tyrosine kinase domain (FLT3-TKD) are found in ~ 8% of patients with AML, with D835Y as the most common substitution. This mutation activates survival signals that drives the disease and is resistant to the first generation FLT3 inhibitors. Development of a highly sensitive method to detect FLT3D835Y is important to direct therapeutic options, predict prognosis, and monitor minimal residual disease in patients with AML. Methods and results In the present study, we developed a highly sensitive FLT3D835Y detection method by using the restriction fragment nested allele-specific PCR technique. The method consists of three steps: 1) initial amplification of DNA samples with PCR primers surrounding the FLT3D835Y mutation site, 2) digestion of the PCR products with restriction enzyme EcoRV that only cleaves the wild type allele, and 3) detection of FLT3D835Y by allele-specific PCR with nested primers. We were able to detect FLT3D835Y with a sensitivity of 0.001% by using purified plasmid DNAs and blood cell DNAs containing known proportions of FLT3D835Y. We analyzed blood cell DNA samples from 64 patients with AML and found six FLT3D835Y-positive cases, two of which could not be detected by conventional DNA sequencing methods. Importantly, the method was able to detect FLT3D835Y in a sample collected from a relapsed patient while the patient was in complete remission with negative MRD determined by flow cytometry. Therefore, our RFN-AS-PCR detected MRD after treatment that was missed by flow cytometry and Sanger DNA sequencing, by conventional methods. Conclusions We have developed a simple and highly sensitive method that will allow for detection of FLT3D835Y at a very low level. This method may have major clinical implications for treatment of AML.
- Published
- 2020
- Full Text
- View/download PDF
30. Thrombopoietin is required for full phenotype expression in a JAK2V617F transgenic mouse model of polycythemia vera
- Author
-
Wanke Zhao, Akil Merchant, Donna M. Williams, Jerry L. Spivak, Linda S. Resar, Alison R. Moliterno, Zhizhuang Joe Zhao, Ophelia Rogers, and Amy S. Duffield
- Subjects
0301 basic medicine ,Male ,Physiology ,Neutrophils ,Hematologic Cancers and Related Disorders ,White Blood Cells ,Mice ,0302 clinical medicine ,Polycythemia vera ,Animal Cells ,hemic and lymphatic diseases ,Immune Physiology ,Medicine and Health Sciences ,Polycythemia Vera ,Thrombocytosis ,Multidisciplinary ,Stem Cells ,Hematopoietic stem cell ,Animal Models ,Hematology ,Body Fluids ,Haematopoiesis ,Phenotypes ,medicine.anatomical_structure ,Blood ,Phenotype ,Experimental Organism Systems ,Oncology ,Thrombopoietin ,030220 oncology & carcinogenesis ,Medicine ,Stem cell ,Cellular Types ,Anatomy ,Receptors, Thrombopoietin ,Research Article ,Thrombocythemia, Essential ,Genetically modified mouse ,Platelets ,Science ,Immune Cells ,Immunology ,Mouse Models ,Mice, Transgenic ,Biology ,Research and Analysis Methods ,Blood Plasma ,03 medical and health sciences ,Model Organisms ,medicine ,Genetics ,Animals ,Humans ,Myelofibrosis ,Thrombopoietin receptor ,Blood Cells ,Myeloproliferative Disorders ,Biology and Life Sciences ,Cancers and Neoplasms ,Cell Biology ,Janus Kinase 2 ,medicine.disease ,Hematopoietic Stem Cells ,Mice, Inbred C57BL ,Disease Models, Animal ,030104 developmental biology ,Primary Myelofibrosis ,Mutation ,Cancer research ,Animal Studies ,Spleen - Abstract
The myeloproliferative neoplasms, polycythemia vera, essential thrombocytosis and primary myelofibrosis are hematopoietic stem cell disorders and share driver mutations that either directly activate the thrombopoietin receptor, MPL, or activate it indirectly through gain-of-function mutations in the gene for JAK2, its cognate tyrosine kinase. Paradoxically, MPL surface expression in hematopoietic stem cells is also reduced in the myeloproliferative neoplasms due to abnormal post-translational glycosylation and premature destruction of JAK2, suggesting that the myeloproliferative neoplasms are disorders of MPL processing since MPL is the only hematopoietic growth factor receptor in hematopoietic stem cells. To examine this possibility, we genetically manipulated MPL expression and maturation in a JAK2V617F transgenic mouse model of polycythemia vera. Elimination of MPL expression completely abrogated the polycythemia vera phenotype in this JAK2V617F transgenic mouse model, which could only be partially restored by expression of one MPL allele. Most importantly, elimination of thrombopoietin gene expression abrogated the polycythemia vera phenotype in this JAK2V617F transgenic mouse model, which could be completely restored by expression of a single thrombopoietin allele. These data indicate that polycythemia vera is in part a thrombopoietin-dependent disorder and that targeting the MPL-thrombopoietin axis could be an effective, nonmyelotoxic therapeutic strategy in this disorder.
- Published
- 2020
31. Finding new lanes: Chimeric antigen receptor (CAR) T‐cells for myeloid leukemia
- Author
-
Zhizhuang Joe Zhao and Suraj Pratap
- Subjects
Cancer Research ,Myeloid ,medicine.medical_treatment ,Sialic Acid Binding Ig-like Lectin 3 ,Interleukin-3 Receptor alpha Subunit ,Review ,Disease ,Immunotherapy, Adoptive ,Antigen ,Leukemia, Myelogenous, Chronic, BCR-ABL Positive ,hemic and lymphatic diseases ,medicine ,Humans ,Chemotherapy ,Receptors, Chimeric Antigen ,business.industry ,Myeloid leukemia ,Chimeric antigen receptor ,Leukemia, Myeloid, Acute ,Haematopoiesis ,medicine.anatomical_structure ,Oncology ,NK Cell Lectin-Like Receptor Subfamily K ,Cancer research ,Bone marrow ,business - Abstract
Background Myeloid leukemia represents a heterogeneous group of cancers of blood and bone marrow which arise from clonal expansion of hematopoietic myeloid lineage cells. Acute myeloid leukemia (AML) has traditionally been treated with multi-agent chemotherapy, but conventional therapies have not improved the long-term survival for decades. Chronic myeloid leukemia (CML) is an indolent disease which requires lifelong treatment, is associated with significant side effects, and carries a risk of progression to potentially lethal blast crises. Recent findings Recent advances in molecular biology, virology, and immunology have enabled researchers to grow and modify T lymphocytes ex-vivo. Chimeric antigen receptor (CAR) T-cell therapy has been shown to specifically target cells of lymphoid lineage and induce remission in acute lymphoblastic leukemia (ALL) patients. While the success of CAR T-cells against ALL is considered a defining moment in modern oncology, similar efficacy against myeloid leukemia cells remains elusive. Over the past 10 years, numerous CAR T-cells have been developed that can target novel myeloid antigens, and many clinical trials are finally starting to yield encouraging results. In this review, we present the recent advances in this field and discuss strategies for future development of myeloid targeting CAR T-cell therapy. Conclusions The field of CAR T-cell therapy has rapidly evolved over the past few years. It represents a radically new approach towards cancers, and with continued refinement it may become a viable therapeutic option for patients of acute and chronic myeloid leukemia.
- Published
- 2020
- Full Text
- View/download PDF
32. Proapoptotic Mitochondrial Carrier Homolog Protein PSAP Mediates Death Receptor 6 Induced Apoptosis
- Author
-
Fuqiang Zhang, Yunzhou Dong, Xueqi Fu, Chen Hu, Jingtian Zhang, Xuemin Xu, Mei-Zhen Cui, Zhizhuang Joe Zhao, Linlin Zeng, and Han Niu
- Subjects
0301 basic medicine ,Apoptosis ,Mitochondrion ,Receptors, Tumor Necrosis Factor ,Mitochondrial Proteins ,03 medical and health sciences ,0302 clinical medicine ,Two-Hybrid System Techniques ,medicine ,Humans ,Receptor ,bcl-2-Associated X Protein ,Gene knockdown ,biology ,Chemistry ,Cell growth ,General Neuroscience ,Cytochrome c ,Neurotoxicity ,Membrane Proteins ,General Medicine ,Mitochondrial carrier ,medicine.disease ,Cell biology ,Mitochondria ,Psychiatry and Mental health ,Clinical Psychology ,030104 developmental biology ,Gene Knockdown Techniques ,biology.protein ,Geriatrics and Gerontology ,030217 neurology & neurosurgery ,HeLa Cells - Abstract
Presenilin-associated protein (PSAP) was originally identified as a mitochondrial proapoptotic protein. To further explore the apoptotic pathway that involves PSAP, our yeast two-hybrid screen revealed that PSAP interacts with a death receptor, DR6. DR6 is a relatively less common member of the death receptor family and has been shown to mediate the neurotoxicity of amyloid-β, mutant SOD1, and prion proteins and has also been implicated in the regulation of immune cell proliferation and differentiation. Our previous study showed that DR6 induces apoptosis via a unique mitochondria-dependent pathway different from the conventional death receptor-mediated extrinsic apoptotic pathways. Thus, the interaction of DR6 with PSAP established a direct molecular link between DR6 and mitochondrial apoptotic pathway. We investigated the possible role of PSAP in DR6-induced apoptosis. Interestingly, it was discovered that knockdown of PSAP strongly inhibited DR6-induced apoptosis. To further elucidate the mechanism by which PSAP mediates DR6-induced mitochondria-dependent apoptosis, our data demonstrated that knockdown of PSAP blocked DR6-induced Bax translocation and cytochrome c release from the mitochondria. Moreover, it was found that both PSAP and DR6 form complexes with Bax, but at different subcellular locations. The DR6-Bax complex was detected in the cytosolic fraction while the PSAP-Bax complex was detected in the mitochondrial fraction. The observation that knockdown of DR6 significantly reduced the amount of PSAP-Bax complex detected in mitochondria suggests a possibility that DR6-bound Bax is transferred to PSAP upon interaction with PSAP at the mitochondria, leading to cytochrome c release and eventually apoptosis.
- Published
- 2020
33. MiR-543 regulates the epigenetic landscape of myelofibrosis by targeting TET1 and TET2
- Author
-
Wanting Tina Ho, Linda Fabris, Elizabeth Torres-Claudio, Roxana S. Redis, Xinna Zhang, Srdan Verstovsek, Cristina Ivan, Taghi Manshouri, Pranav Narayanan, Nayra Soares do Amaral, Masayoshi Shimizu, Zhizhuang Joe Zhao, Geoffrey Bartholomeusz, Cristina Perez, Enrique Fuentes-Mattei, Leonard Golfman, Pilar Mur, Adriana Badillo-Perez, Mihnea P. Dragomir, Erik Knutsen, Patrick A. Zweidler-McKay, Zeev Estrov, Andreia M. Silva, Marcos Roberto Estecio, Ioana Berindan-Neagoe, Ciprian Tomuleasa, Diana Gulei, Recep Bayraktar, Wanke Zhao, George A. Calin, and Instituto de Investigação e Inovação em Saúde
- Subjects
0301 basic medicine ,Ruxolitinib ,DNA-Binding Proteins / genetics ,Myelofibrosis ,Epigenesis, Genetic ,DNA-Binding Proteins / drug effects ,Mixed Function Oxygenases ,Histones ,Mice ,0302 clinical medicine ,Janus Kinase Inhibitors / therapeutic use ,Janus Kinases / metabolism ,Primary Myelofibrosis / genetics ,Hematology ,Primary Myelofibrosis / drug therapy ,General Medicine ,MicroRNAs / metabolism ,Extramedullary hematopoiesis ,DNA-Binding Proteins ,MicroRNAs / pharmacology ,030220 oncology & carcinogenesis ,Cytokines ,medicine.drug ,Research Article ,STAT3 Transcription Factor ,medicine.medical_specialty ,Proto-Oncogene Proteins / drug effects ,Dioxygenases ,03 medical and health sciences ,Downregulation and upregulation ,Internal medicine ,Proto-Oncogene Proteins ,Hematopoesi ,Nitriles ,medicine ,Janus Kinase Inhibitors ,Animals ,Humans ,Epigenetics ,Proto-Oncogene Proteins / genetics ,Protein Kinase Inhibitors ,Myeloproliferative neoplasm ,Janus Kinases ,Cytopenia ,Myeloproliferative Disorders ,Mielofibrosi ,business.industry ,Protein Kinase Inhibitors / pharmacology ,Pyrazoles / therapeutic use ,medicine.disease ,United States ,Hematopoiesis ,MicroRNAs ,Disease Models, Animal ,Epigenesis, Genetic / drug effects ,030104 developmental biology ,Cytokines / metabolism ,MicroRNAs / genetics ,Pyrimidines ,Primary Myelofibrosis ,Mutation ,Cancer research ,Pyrazoles ,business ,Transcriptome - Abstract
Myelofibrosis (MF) is a myeloproliferative neoplasm characterized by cytopenia and extramedullary hematopoiesis, resulting in splenomegaly. Multiple pathological mechanisms (e.g., circulating cytokines and genetic alterations, such as JAKV617F mutation) have been implicated in the etiology of MF, but the molecular mechanism causing resistance to JAK2V617F inhibitor therapy remains unknown. Among MF patients who were treated with the JAK inhibitor ruxolitinib, we compared noncoding RNA profiles of ruxolitinib therapy responders versus nonresponders and found miR-543 was significantly upregulated in nonresponders. We validated these findings by reverse transcription–quantitative PCR. in this same cohort, in 2 additional independent MF patient cohorts from the United States and Romania, and in a JAK2V617F mouse model of MF. Both in vitro and in vivo models were used to determine the underlying molecular mechanism of miR-543 in MF. Here, we demonstrate that miR-543 targets the dioxygenases ten-eleven translocation 1 (TET1) and 2 (TET2) in patients and in vitro, causing increased levels of global 5-methylcytosine, while decreasing the acetylation of histone 3, STAT3, and tumor protein p53. Mechanistically, we found that activation of STAT3 by JAKs epigenetically controls miR-543 expression via binding the promoter region of miR-543. Furthermore, miR-543 upregulation promotes the expression of genes related to drug metabolism, including CYP3A4, which is involved in ruxolitinib metabolism. Our findings suggest miR-543 as a potentially novel biomarker for the prognosis of MF patients with a high risk of treatment resistance and as a potentially new target for the development of new treatment options The work in GAC’s laboratory was partly supported by NIH/National Center for Advancing Translational Sciences grant UH3TR00943-01, the NIH/National Cancer Institute (NCI) grant 1 R01 CA182905-01, U54 grant UPR/MDACC Partnership for Excellence in Cancer Research 2016 Pilot Project, Team Department of Defense grant CA160445P1, a Ladies Leukemia League grant, a Chronic Lymphocytic Leukemia Moonshot Flagship project, a Sister Institution Network Fund 2017 grant, and the Estate of C. G. Johnson, Jr. EFM was supported in part by award number P50 CA140388 from the NCI and by the NIH Clinical Research Loan Repayment Program. AMS was supported by Fundação para a Ciência e a Tecnologia through fellowship SFRH/BD/85968/2012. ZJZ’s work was supported by grants from the MPN Foundation and the Oklahoma Center for the Advancement of Science and Technology. DG, MPD, and IBN were supported in part by a Programul Opera¿ional Competitivitate grant nr35/01.09.2016, ID 37_796, titled “Clinical and economical impact of personalized targeted anti-mi-croRNA therapies in reconverting lung cancer chemoresistance” (CANTEMIR). NSDA was supported by the Fundação de Amparo à Pesquisa do Estado de São Paulo, BEPE 2016/09349-4.
- Published
- 2020
34. Structure analysis of the receptor binding of 2019-nCoV
- Author
-
Zhizhuang Joe Zhao, Yao Guo, Yihang Pan, and Yun Chen
- Subjects
0301 basic medicine ,Molecular model ,Sequence analysis ,Biophysics ,Phenylalanine ,medicine.disease_cause ,Biochemistry ,Virus ,03 medical and health sciences ,0302 clinical medicine ,medicine ,Molecular Biology ,Coronavirus ,chemistry.chemical_classification ,biology ,Cell Biology ,Cell biology ,Amino acid ,COVID-19 ,Structure analysis ,Molecular modeling ,030104 developmental biology ,chemistry ,030220 oncology & carcinogenesis ,biology.protein ,Antibody ,Glycoprotein ,hormones, hormone substitutes, and hormone antagonists - Abstract
2019-nCoV is a newly identified coronavirus with high similarity to SARS-CoV. We performed a structural analysis of the receptor binding domain (RBD) of spike glycoprotein responsible for entry of coronaviruses into host cells. The RBDs from the two viruses share 72% identity in amino acid sequences, and molecular simulation reveals highly similar ternary structures. However, 2019-nCoV has a distinct loop with flexible glycyl residues replacing rigid prolyl residues in SARS-CoV. Molecular modeling revealed that 2019-nCoV RBD has a stronger interaction with angiotensin converting enzyme 2 (ACE2). A unique phenylalanine F486 in the flexible loop likely plays a major role because its penetration into a deep hydrophobic pocket in ACE2. ACE2 is widely expressed with conserved primary structures throughout the animal kingdom from fish, amphibians, reptiles, birds, to mammals. Structural analysis suggests that ACE2 from these animals can potentially bind RBD of 2019-nCoV, making them all possible natural hosts for the virus. 2019-nCoV is thought to be transmitted through respiratory droplets. However, since ACE2 is predominantly expressed in intestines, testis, and kidney, fecal-oral and other routes of transmission are also possible. Finally, antibodies and small molecular inhibitors that can block the interaction of ACE2 with RBD should be developed to combat the virus.
- Published
- 2020
35. Phenotype risk scores identify patients with unrecognized Mendelian disease patterns
- Author
-
Wanting T. Ho, Lisa Bastarache, Dan M. Roden, Andrew M. Glazer, Jacob J. Hughey, Nancy J. Cox, Sara L. Van Driest, Michael Temple, Wanke Zhao, Joy D. Cogan, Scott J. Hebbring, Joshua C. Denny, Jonathan D. Mosley, Murray H. Brilliant, Rizwan Hamid, Joy E. Marlo, Travis J. Osterman, Wei-Qi Wei, QiPing Feng, Digna R. Velez Edwards, Tracy L. McGregor, Robert J. Carroll, Zhizhuang Joe Zhao, Todd L. Edwards, Andrea H. Ramirez, Quinn S. Wells, and Douglas M. Ruderfer
- Subjects
0301 basic medicine ,DNA Mutational Analysis ,Biology ,Mendelian disease ,Article ,03 medical and health sciences ,symbols.namesake ,Risk Factors ,Electronic health record ,Databases, Genetic ,Genetic variation ,Electronic Health Records ,Humans ,Exome ,Genetic Predisposition to Disease ,Genetic Association Studies ,Genetic association ,Genetics ,Multidisciplinary ,Genetic Diseases, Inborn ,Genetic variants ,Genetic Variation ,Phenotype ,030104 developmental biology ,Mendelian inheritance ,symbols - Abstract
Hidden effects of Mendelian inheritance Identifying the determinate factors of genetic disease has been quite successful for Mendelian inheritance of large-effect pathogenic variants. In these cases, two non- or low-functioning genes contribute to disease. However, Mendelian effects of lesser strength have generally been ignored when looking at genomic consequences in human health. Bastarache et al. used electronic records to identify the phenotypic effects of previously unidentified Mendelian variations. Their analysis suggests that individuals with undiagnosed Mendelian diseases may be more prevalent in the general population than assumed. Because of this, genetic analysis may be able to assist clinicians in arriving at a diagnosis. Science , this issue p. 1233
- Published
- 2018
- Full Text
- View/download PDF
36. 66 The association between increasing patient body mass index and gynecologic operating room scheduling inaccuracies
- Author
-
K. Chaves, T. Ding, A. Yunker, and Zhizhuang Joe Zhao
- Subjects
medicine.medical_specialty ,business.industry ,Association (object-oriented programming) ,Emergency medicine ,medicine ,Scheduling (production processes) ,Obstetrics and Gynecology ,business ,Body mass index - Published
- 2021
- Full Text
- View/download PDF
37. 98 The association between bariatric surgery and need for fibroid procedural intervention amongst women with fibroids
- Author
-
H. Curlin, K.F. Chaves, Lfb Harvey, D.R. Velez Edwards, and Zhizhuang Joe Zhao
- Subjects
medicine.medical_specialty ,business.industry ,Intervention (counseling) ,General surgery ,Obstetrics and Gynecology ,Medicine ,business ,Association (psychology) - Published
- 2021
- Full Text
- View/download PDF
38. FLT3 Inhibitors Induce p53 Instability Due to Increased Interaction between p53 and MDM2 By Inhibiting STAT5 Phosphorylation in Acute Myeloid Leukemia
- Author
-
Liuting Yu, Zhizhuang Joe Zhao, Ming Yang, Yuming Zhao, Xiaojun Xu, Han Zhong Pei, Xiaomei Zhuang, Bo Lu, Zhiguang Chang, Dengyang Zhang, Yun Chen, and Yao Guo
- Subjects
biology ,Chemistry ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,Biochemistry ,Instability ,hemic and lymphatic diseases ,biology.protein ,Cancer research ,Phosphorylation ,Mdm2 ,STAT5 - Abstract
Frequently mutated in Acute myeloid leukemia (AML), FLT3 is considered as one of the favorable targets for treatment. The FLT3 internal tandem duplication (ITD) mutation enhances kinase activity and causes hyperactivation of downstream signal transduction. Several small molecule FLT3 inhibitors have developed, but their clinical efficacy is limited due to generation of drug resistance. In this study, we define a new mechanism of drug resistance toward tyrosine kinase inhibitors (TKIs). Initially, we found a rapid decrease in the protein level of tumor suppressor p53 in FLT3-ITD-positive MV4-11 and MOLM13 cells and peripheral blood mononuclear cells (PBMCs) from FLT3-ITD AML patients upon treatment with TKIs including sorafenib, sunitinib and quizartinib. The decrease is not caused by changes in mRNA expression as revealed by qPCR analyses but rather by accelerated protease degradation because the p53 protein was stabilized by proteasome inhibitor MG132. Furthermore, treatment of cells with RG7388, a potent disruptor of p53 and MDM2 interaction, prevented the TKI-induced p53 loss. Since MDM2 is the most important E3 ligase responsible for ubiquitination of p53, the data suggest that TKIs may lead to the degradation of p53 by promoting ubiquitination. Indeed, ubiquitination assays verified that TKIs promoted K48 poly-ubiquitination of p53. Previous studies have demonstrated that activations of FLT3 downstream signaling components such as ERKs and Akt reduce p53 protein stability through ubiquitination by activating MDM2. It is somewhat unexpected that inhibition of FLT3-ITD and its downstream signaling pathways also resulted in decreased p53 stability due to increased ubiquitination. We treated FLT3-ITD-containing cells with specific ERK, AKT and STAT5 inhibitors. Interestingly, while inhibition of ERKs and AKT had no significant effect on the stability of p53, STAT5 inhibition resulted in a reduced level of p53 accompanied by increased K48 poly-ubiquitination. We further analyzed the interaction of p53 with MDM2 in AML cells by using immunoprecipitation. The results showed that the p53-MDM2 interaction was significantly enhanced after treatment with TKIs and STAT5 inhibitors, which was diminished in the presence of RG7388. Subcellular fractionation revealed the presence of p53 and STAT5 in both nucleus and cytoplasm. Treatment of cells with TKIs resulted in a decreased level of p53 and STAT5 in the nucleus, and immunoprecipitation of nuclear proteins with a p53 antibody revealed a reduced association of p53 with STAT5. Taken together, the data suggest that FLT3 inhibitors inhibited nuclear translocation of STAT5 and reduced its interaction of p53 thereby facilitating p53/MDM2 interaction and subsequent ubiquitination and degradation of p53. This study reveals a novel mechanism by which drug resistance to TKIs may occur and further support the use of MDM2/p53 interaction inhibitors in combination with TKIs for treatment of AML. Disclosures No relevant conflicts of interest to declare.
- Published
- 2021
- Full Text
- View/download PDF
39. HMGA1 Chromatin Regulators Drive Progression in Myeloproliferative Neoplasms through Epigenetic Rewiring to Induce Networks Involved in GATA2 and Proliferation
- Author
-
Jerry L. Spivak, Liping Li, Matthew C. Stubbs, Donna M. Williams, Raajit K. Rampal, Ophelia Rogers, Marija Vasiljevic, Leslie Cope, Alison R. Moliterno, Karen L. Reddy, Richard Koche, Daniel R. Matson, Joseph Kim, Jung-Hyun Kim, Wenyan Lu, Linda M.S. Resar, Lingling Xian, Li Luo, and Zhizhuang Joe Zhao
- Subjects
Immunology ,GATA2 ,biology.protein ,Cell Biology ,Hematology ,Epigenetics ,Biology ,Biochemistry ,HMGA1 ,Cell biology ,Chromatin - Abstract
Introduction: Myeloproliferative neoplasms (MPN) are clonal hematopoietic stem cell (HSC) disorders characterized by hyperactive JAK/STAT signaling and increased risk of transformation to myelofibrosis (MF) and acute myeloid leukemia (AML). However, mechanisms driving progression remain elusive and therapies are ineffective after leukemic transformation. The High Mobility Group A1 (HMGA1) gene encodes oncogenic chromatin regulators which are overexpressed in diverse tumors where they portend adverse outcomes (Resar Cancer Res 2010; Xian et al Nature Commun 2017). Hmga1 induces leukemic transformation in transgenic mice and HMGA1 is overexpressed in refractory myeloid malignancies (Resar et al Cancer Res 2018). Further, germline lesions within the HMGA1 loci increase the risk for developing MPN (Bao et al Nature 2020). We therefore sought to: 1) test the hypothesis that HMGA1 drives MPN progression by rewiring transcriptional networks to foster leukemogenesis, and, 2) identify mechanisms underlying HMGA1 that could be targeted with therapy. Methods: To elucidate the function of HMGA1, we disrupted HMGA1 expression via CRISPR/Cas9 or short hairpin RNA (shRNA) targeting 2 different sequences per gene and assessed proliferation, colony formation, apoptosis, and leukemogenesis. We also generated JAK2 V617F transgenic mouse models of MF with Hmga1 deficiency. To dissect molecular mechanisms underlying HMGA1, we integrated RNAseq, ATACseq, and chromatin immunoprecipitation (ChIP) from MPN-AML cell lines (DAMI, SET-2). Next, we tested whether HMGA1 depletion synergizes with ruxolitinib in preventing leukemic engraftment in mice. To identify drugs to target HMGA1 networks, we applied the Broad Institute Connectivity Map (CMAP). Results: HMGA1 is overexpressed in CD34 + cells from patients with JAK2 V617F MPN with highest levels after transformation to MF or AML in 3 independent cohorts. CRISPR/Cas9 inactivation or shRNA-mediated HMGA1 silencing disrupts proliferation, decreases the frequency of cells in S phase, increases apoptosis, and impairs clonogenicity in human MPN-AML cell lines. HMGA1 depletion also prevents leukemic engraftment in mice. Surprisingly, loss of just a single Hmga1 allele prevents progression to MF in JAK2 V617Fmurine models of MPN, decreasing erythrocytosis, thrombocytosis, and preventing splenomegaly and fibrosis of the spleen and bone marrow. Further, Hmga1 deficiency preferentially prevents expansion in long-term HSC, granulocyte-macrophage progenitors, and megakaryocyte-erythroid progenitors in JAK2 V617F mice. RNAseq revealed genes induced by HMGA1 that govern cell cycle progression (E2F targets, mitotic spindle, G2M checkpoint, MYC targets) and cell fate decisions (GATA2 networks), including the GATA2 master regulator gene. Silencing GATA2 recapitulates anti-leukemia phenotypes observed with HMGA1 deficiency whereas restoring GATA2 in MPN-AML cells with HMGA1 silencing partially rescues leukemia phenotypes, increasing clonogenicity and leukemic engraftment. Mechanistically, HMGA1 binds directly to AT-rich sequences near the GATA2 developmental enhancer (+9.5), enhances chromatin accessibility, and recruits active histone marks (H3K4me1/3) to induce GATA2 expression. HMGA1 depletion enhances responses to the JAK/STAT Inhibitor, ruxolitinib, delaying leukemic engraftment and prolonging survival in murine models of JAK2 V617F MPN-AML. Further, epigenetic drugs predicted to target HMGA1 transcriptional networks using CMAP synergize with JAK inhibitors to disrupt proliferation in human MPN-AML cells. HMGA1 and GATA2 are co-expressed and up-regulated with progression from MF to AML in matched patient samples. Moreover, HMGA1 transcriptional networks are activated in leukemic blasts, thus underscoring the role of HMGA1 in human MPN progression. Conclusions: We uncovered a previously unknown epigenetic program whereby HMGA1 enhances chromatin accessibility and recruits activating histone marks to induce transcriptional networks required for progression in MPN, including direct transactivation of GATA2. Further, HMGA1 networks can be targeted with epigenetic therapy and synergize with ruxolitinib. Together, our studies reveal a new paradigm whereby HMGA1 up-regulates GATA2 and proliferation networks to drive disease progression and illuminate HMGA1 as a novel therapeutic target in MPN. Figure 1 Figure 1. Disclosures Rampal: Jazz Pharmaceuticals: Consultancy; Incyte: Consultancy, Research Funding; Kartos: Consultancy; Constellation: Research Funding; Pharmaessentia: Consultancy; Blueprint: Consultancy; Disc Medicine: Consultancy; Stemline: Consultancy, Research Funding; BMS/Celgene: Consultancy; Novartis: Consultancy; Sierra Oncology: Consultancy; CTI: Consultancy; Abbvie: Consultancy; Memorial Sloan Kettering: Current Employment. Stubbs: Incyte Research Institute: Current Employment, Current holder of individual stocks in a privately-held company.
- Published
- 2021
- Full Text
- View/download PDF
40. Small Molecule Kinase Inhibitor Sunitinib Specifically Targets Juvenile Myelomonocytic Leukemia Cells with SHP2(PTPN11) Mutation
- Author
-
Zhizhuang Joe Zhao, Yuming Zhao, Junbin Huang, Yun Chen, Chunxiao He, Chun Chen, Hongman Xue, and Yao Guo
- Subjects
Juvenile myelomonocytic leukemia ,Kinase ,Chemistry ,Sunitinib ,Immunology ,Cell Biology ,Hematology ,medicine.disease ,Biochemistry ,Small molecule ,PTPN11 ,Mutation (genetic algorithm) ,Cancer research ,medicine ,medicine.drug - Abstract
Juvenile myelomonocytic leukemia (JMML) is a highly fatal malignant disease in early childhood. It is still unknown of the specific pathogenesis, and there is shortage of effective targeted therapeutic approaches. Gain of function SHP2 mutation encoded by PTPN11 gene is found in approximately 35% of JMML patients, which maybe contributed to its pathogenesis. JMML patients with SHP2 mutation have lower survival rate and higher recurrence rate. All of the above make development of new therapies imperative. Currently, there is no stable cell line that can accurately reflect the characteristics of JMML abnormal cells for research on JMML. In this study, we established two leukemia cell lines that depend on mutated SHP2 for survival, and discovered promising drugs that targeted mutated-SHP2-dependent oncogenic signaling pathway through drug screening method. HCD-57 cells are murine erythroleukemia cells that solely depend on exogenic erythropoietin (EPO) for survival. We constructed SHP2-D61Y and SHP2-E76K transformed HCD-57 cell lines through retroviral vectors, the survival of which dependent on mutated SHP2 mediated signaling pathway. Based on these cells, we established a drug screening platform and screened small molecule compound library containing 2862 FDA-approved drugs and 1707 kinase inhibitors. We performed cell viability, flow cytometry, Wright-Giemsa staining, and western blot to evaluate cells after drug treatment. To further assess therapeutic potential, we established in-vivo transplantation model that SHP2-D61Y transformed HCD-57 cells were implanted into immunodeficient NCG mice, and verified the effectiveness of the in-vitro screened drugs. We found that the survival and proliferation of HCD-57 cells transduced by SHP2-D61Y and SHP2-E76K no longer required EPO, but completely relied on the abnormal activation of signaling pathway mediated by mutated SHP2. Western blot results showed that the phosphorylation status of ERK1/2 and AKT of HCD-57 cells expressing SHP2 mutation were abnormally increased, consistent with SHP2-mutated JMML. Thus, we have obtained the leukemia cell lines that can represent the characteristics of activated signaling pathway in JMML with SHP2 mutation. Through drug screening, we observed that drug sunitinib (Sutent ®) selectively inhibits SHP2-mutated HCD-57 cell lines. CCK-8-based cell viability assay demonstrated a dose-dependent inhibition of SHP2-D61Y and SHP2-E76K transformed HCD-57 cell and no effects on the parental HCD-57 cells. Live cell counting with trypan blue revealed that the proliferation of SHP2-mutated HCD-57 cells was totally halted after one day upon treatment with 250 nM sunitinib, whereas the HCD-57 cells were unaffected. Wright-Giemsa staining demonstrated that SHP2-mutated HCD-57 cells showed no normal morphology change and no mitotic activity under sunitinib treatment, otherwise parental HCD-57 cells showed normal mitotic activity. Sunitinib induced apoptosis and cell cycle arrest at G1 phase in SHP2-mutated HCD-57 cells by flow cytometry, but had little effect on the parental HCD-57 cells. Sunitinib effectively downregulates the phosphorylation of ERK and AKT in SHP2-mutated cells, revealing the mechanism of sunitinib targeting SHP2-mutated cells. In addition, after transplantation of SHP2-D61Y transformed HCD-57 cells for 3 weeks, the spleen of NCG mice increased from an average of 45 mg to more than 300 mg; flow cytometry analysis showed that the implanted cells accounted for over 75% of the total nucleated cells in the bone marrow and spleen. Compared with the vehicle control, the number of monocytes in these mice was reduced to the normal range by treatment with sunitinib, and the spleen weights were reduced by about 50%. Histochemical staining showed disappearance of the myeloid infiltration in the spleen, liver and bone marrow. The above results all indicate that sunitinib has strong in-vivo anti-leukemia activity. Furthermore, western blot analysis showed that the administration of sunitinib significantly inhibited the phosphorylation expression level of AKT and ERK, indicating the effectivity of sunitinib in vivo. In conclusion, our data demonstrated that HCD-57 cell line is an effective tool for studying oncogenic signaling pathway and screening drugs that targeted JMML with SHP2 mutation. Sunitinib can be an effective drug for the targeted treatment of JMML in the future. Disclosures No relevant conflicts of interest to declare.
- Published
- 2021
- Full Text
- View/download PDF
41. Establishment of Stable Cell Lines Representing Juvenile Myelomonocytic Leukemia with SHP2 Mutations
- Author
-
Yao Guo, Ming Yang, Liuting Yu, Dengyang Zhang, Junbin Huang, Zhiguang Chang, Zhizhuang Joe Zhao, Yun Chen, Yuming Zhao, Han Zhong Pei, Chun Chen, Chunxiao He, and Hongman Xue
- Subjects
Juvenile myelomonocytic leukemia ,Cell culture ,Immunology ,Cancer research ,medicine ,Cell Biology ,Hematology ,Biology ,medicine.disease ,Biochemistry - Abstract
Protein tyrosine phosphatase SHP2 encoded by PTPN11 is a key regulator in growth factor and cytokine signaling. Overwhelming evidence suggests its vital role in hematopoietic stem cell function and hematopoiesis. As a bona fide proto-oncogene product, gain-of-function mutations of SHP2 cause hematological malignancies, most notably juvenile myelomonocytic leukemia (JMML) which bear somatic SHP2 mutations in 35% of cases. Numerous studies have utilized murine models to investigate the role of mutant SHP2 in hematopoiesis and leukemogenesis and successfully produced resembling myeloproliferative neoplasm (MPN) and even full-blown leukemia in recipient animals. However, mutant SHP2-transformed cell lines have not been generated. In the present study, we established oncogenic mutant SHP2-transformed cell lines from erythropoietin (EPO)-dependent HCD-57 erythroid leukemia cells. First, we generated recombinant retroviruses expressing SHP2-D61Y and SHP2-E76K, the two most common SHP2 mutants found in individuals with JMML, by using the pMSCV-IRES-GFP vector. We then infected HCD-57 cells with the recombinant retroviruses. Unlike the parent HCD-57 cells, the infected cells were able to grow in the absence of EPO as demonstrated by viable GFP-positive cells. We further performed semi-solid methylcellulose colony cultures and isolated single clones of EPO-independent HCD57 cells. The isolated clonal cells overexpressed mutant SHP2 and proliferate rapidly in the absence of EPO. In contrast, HCD57 cells infected with retroviruses expressing wild type SHP2 failed to survive in the absence of EPO, indicating only gain-of-function mutant forms of SHP2 have the cell-transformation capability. We also carried out parallel experiments with the pro-B Ba/F3 cell line that require interleukin 3 (IL3) for survival. Interestingly, over-expression of SHP2-D61Y and SHP2-E76K was not sufficient to give rise to IL3-indepdent Ba/F3 cells, suggesting that HCD57 cells have some unique properties making them susceptible for transformation by oncogenic SHP2 mutants. We further performed in vitro and in vivo characterization of transformed HCD57 cells. Cell signaling analyses revealed that both HCD57-SHP2-D61Y and HCD57-SHP2-E76Kcells exhibited aberrantly elevated levels of pERK and pAKT in the absence of cytokine stimulation, which was consistent with the notion that gain-of-function SHP2 mutants perturb growth control through deregulation of the Ras signaling pathway. Upon intravenous injection into immunodeficient mice, the SHP2 mutant-transformed HCD57 cells caused acute leukemia with markedly increased spleen. Finally, we screened a small molecule inhibitor library to identify compounds that may specifically target the SHP2 mutants. We found several tyrosine kinase inhibitors including dasatinib and trametinib potently inhibited HCD57-SHP2-D61Y and HCD57-SHP2-E76Kcells but not the parent HCD57 cells. At sub-micromolar concentrations, dasatinib and trametinib abolished elevated ERK and Akt activation caused by the SHP2 mutants. This study not only proves that gain-of function mutations of SHP2 are capable of fully transforming cells but also provides a unique cell system to study pathogenesis of SHP2 mutants and to identify specific inhibitors for drug development. Disclosures No relevant conflicts of interest to declare.
- Published
- 2021
- Full Text
- View/download PDF
42. The Association between Patient Body Mass Index and Initial Procedure Performed as Treatment of Fibroids
- Author
-
H. Curlin, Zhizhuang Joe Zhao, D.R. Velez Edwards, L.F.B. Harvey, and K.F. Chaves
- Subjects
medicine.medical_specialty ,Hysterectomy ,business.industry ,Uterine fibroids ,Obstetrics ,medicine.medical_treatment ,Obstetrics and Gynecology ,Retrospective cohort study ,Overweight ,Logistic regression ,medicine.disease ,Obesity ,female genital diseases and pregnancy complications ,Obstructive sleep apnea ,Medicine ,medicine.symptom ,business ,Body mass index - Abstract
Study Objective To compare the distribution of initial procedures performed as treatment of uterine fibroids between the different classes of patient body mass index (BMI). Design Retrospective cohort study. Setting Academic medical center. Patients or Participants We included patients (N=1982) with known fibroids (based on ICD codes and ultrasound findings) who underwent a fibroid procedural intervention between 2008 and 2015. Interventions Uterine fibroid embolization (UFE), myomectomy (excluding hysteroscopic procedures), or hysterectomy Measurements and Main Results The main exposure was patient BMI class: normal weight (n=498), overweight (n=518), and class I (n=413), II (n=260), and III (n=293) obesity. The main outcome was the first fibroid procedural intervention (UFE, myomectomy, or hysterectomy) performed during the study time period. Amongst women of normal weight who underwent a fibroid procedural intervention, 67.5% of the time this procedure was hysterectomy. Comparatively, overweight women and women with class I, II, and III obesity underwent hysterectomy as their initial procedure 71.4%, 77.7%, 78.1%, and 78.8% of the time (p Multivariable logistic regression adjusted for patient age, race, and the presence of the obesity-related comorbidities of hypertension, diabetes, and obstructive sleep apnea. When compared to women of normal weight, women with class I (aOR 1.8, 95% CI 1.3-2.5, p Conclusion Obese women were significantly more likely to undergo hysterectomy as their initial fibroid procedural intervention as opposed to uterine-preserving procedures, even when adjusted for key factors that may influence a patient's and surgeon's decision regarding operative management. Future studies elaborating on indication for surgery and fibroid burden may shed light on this phenomenon.
- Published
- 2021
- Full Text
- View/download PDF
43. The Relationship between Increasing Patient Body Mass Index and Operating Room Scheduling Inaccuracies at the Time of Hysterectomy
- Author
-
K.F. Chaves, T. Ding, Zhizhuang Joe Zhao, Amanda Yunker, and A. Apple
- Subjects
medicine.medical_specialty ,Hysterectomy ,business.industry ,Obstetrics ,medicine.medical_treatment ,Obstetrics and Gynecology ,Retrospective cohort study ,Overweight ,medicine.disease ,Obesity ,Patient age ,Medicine ,Operative time ,Surgical history ,medicine.symptom ,business ,Body mass index - Abstract
Study Objective To assess the relationship between patient body mass index (BMI) and operating room (OR) scheduling inaccuracies at the time of hysterectomy. Design Retrospective cohort study. Setting Academic medical center. Patients or Participants Scheduled hysterectomies (n=951) performed from 11/2017 through 02/2020 for any gynecologic indication. Interventions Hysterectomy by any route. Measurements and Main Results The primary outcome was operating room time scheduling inaccuracy in minutes, defined as the difference between the scheduled and the actual operating room time. The mean scheduling inaccuracy for women with normal weight, overweight, and class I, II, and III obesity was 22.2, 28.4, 33.5, 38.3, and 48.2 minutes, respectively (p Multivariable linear regression analysis adjusted for patient age, the presence of major cardiopulmonary comorbidities, ASA category, anesthetic type, abdominal surgical history, number of procedures performed, procedure route, and uterine weight. Normal and overweight women were similarly under-scheduled. Compared with normal-weight women, those with class 1 and 3 obesity were, on average, 12.7 (95% CI 2.1-23.3, p=0.019) and 18.9 (95% CI 4.4-33.3, p=0.010), respectively, more minutes under-scheduled. (The average scheduling inaccuracy of women with class 2 obesity did not differ significantly from women with normal weight.) Obese women whose surgeries were under-scheduled, compared to those who were not, spent a significantly greater proportion of their operating room time in operative time (82.6% vs. 74.6%, p Conclusion When adjusted for key factors that may affect operating room time, obese women were significantly more under-scheduled than women of normal weight. Obese women with under-scheduled cases spent proportionally more OR time in operative as opposed to non-operative time. Future studies can assess whether awareness of this tendency helps surgeons more accurately predict operating room time required for hysterectomy for obese women.
- Published
- 2021
- Full Text
- View/download PDF
44. Genetic defects of the IRF1-mediated major histocompatibility complex class I antigen presentation pathway occur prevalently in the JAK2 gene in non-small cell lung cancer
- Author
-
Jie Wu, Tao Shen, Zhizhuang Joe Zhao, and Zhengming Chen
- Subjects
0301 basic medicine ,biology ,MHC class I antigen ,Antigen presentation ,Major histocompatibility complex ,Immune checkpoint ,3. Good health ,03 medical and health sciences ,030104 developmental biology ,0302 clinical medicine ,Immune system ,Oncology ,Interferon ,CDKN2A ,030220 oncology & carcinogenesis ,Immunology ,biology.protein ,medicine ,Cancer research ,Cytotoxic T cell ,medicine.drug - Abstract
Recognition of major histocompatibility complex (MHC) class I antigens on tumor cells by cytotoxic T cells is involved in T cell-mediated tumor immune surveillance and immune checkpoint therapy. The interferon-γ (IFNγ)-IRF1 signaling pathway regulates MHC class I antigen presentation. To examine genetic defects of the IFNγ-IRF1 pathway in non-small cell lung cancer (NSCLC), we analyzed The Cancer Genome Atlas (TCGA) lung adenocarcinoma (LuAd) and lung squamous cell carcinoma (LuSc) data. Loss-of-function (LOF) genetic alterations of the IFNγ-IRF1 pathway genes (IFNGR1, IFNGR2, JAK1, JAK2, STAT1, IRF1) were found in 64 (6.3%) of 1,016 patients. These genetic defects occur prevalently in JAK2 (33 cases) and often through deletions (29 cases) of chromosome 9p24.1. JAK2 deletions were frequently, but not always, associated with deletions of PD-L1 gene (CD274), PD-L2 gene (PDCD1LG2), PTPRD, and CDKN2A/CDKN2B at the chromosome 9p24.1-9p21.3 region. IRF1 expression was correlated with immune cytolytic activity markers GZMA and PRF1 in NSCLC. IFNγ induced IRF1 expression and cell surface HLA-A/HLA-B/HLA-C (HLA-ABC) in A549, H661, H292, and H2172 cells that contained the wildtype JAK2, but not in H1573 and H1623 cells that were JAK2 defective. Deletion of JAK2 or inhibition of the JAK2 kinase activity resulted in loss of IFNγ-induced IRF1 and cell surface HLA-ABC in JAK2 wildtype NSCLC cells, whereas expression of exogenous JAK2 in H1573 cells restored the IFNγ responses. These findings show that JAK2 deficiency is the major mechanism of genetic defects of the IFNγ-IRF1 pathway in NSCLC and reveal a previously unrecognized significance of chromosome 9p deletion in NSCLC.
- Published
- 2017
- Full Text
- View/download PDF
45. Tumor Associated Macrophages Express High-Levels of FLT3 Ligand, Which Induces Activation of FLT3 Signaling That Promotes Survival of Neoplastic Cells in B-Cell Acute Lymphoblastic Leukemia
- Author
-
Yao Guo, Dengyang Zhang, Chun Chen, Xiaojun Xu, Han Zhong Pei, Zhiguang Chang, Zhizhuang Joe Zhao, Hongman Xue, Junbin Huang, Liqin Zhang, Yun Chen, Liuting Yu, and Yuming Zhao
- Subjects
biology ,business.industry ,Lymphoblastic Leukemia ,Immunology ,Cell Biology ,Hematology ,B-cell acute lymphoblastic leukemia ,Malignancy ,medicine.disease ,Biochemistry ,Receptor tyrosine kinase ,Tyrosine Kinase 3 ,biology.protein ,medicine ,Cancer research ,In patient ,Flt3 ligand ,business ,Pathological - Abstract
B-cell acute lymphoblastic leukemia (B-ALL) is a malignancy of B lymphocytes blocked at an early stage of differentiation, which accounts for approximately 30% of childhood cancers. Almost all B-ALL blasts aberrantly express FMS-like tyrosine kinase 3 (FLT3), a receptor tyrosine kinase involved in the development of dendritic cells and B lymphocytes. In B-ALL, the high expression of FLT3 in blasts significantly increases the odds of relapse/death in patients, indicating a pathological role of FLT3 in B-ALL. Note that gain-of-function mutations of FLT3 are relatively rare ( Disclosures No relevant conflicts of interest to declare.
- Published
- 2020
- Full Text
- View/download PDF
46. FLT3 Ligand-DM1 Conjugate Selectively Targets Acute Myeloid Leukemia Cells with FLT3 Expression
- Author
-
Yao Guo, Dengyang Zhang, Zhiguang Chang, Zhizhuang Joe Zhao, Ningning Li, Jian Yu, Yuming Zhao, Chun Chen, Yihang Pan, Han Zhong Pei, Liuting Yu, Chengming Zhu, Hongman Xue, Junbin Huang, and Yun Chen
- Subjects
fluids and secretions ,Chemistry ,hemic and lymphatic diseases ,embryonic structures ,Immunology ,Cancer research ,Myeloid leukemia ,hemic and immune systems ,Flt3 ligand ,Cell Biology ,Hematology ,Biochemistry ,Conjugate - Abstract
Acute myeloid leukemia (AML) is a malignant hematopoietic neoplasm featured by impaired differentiation and uncontrolled proliferation of myeloid progenitors. Gain-of-function mutations of FMS-like tyrosine kinase 3 (FLT3) present in 30-40% of patients with AML. In addition, more than 90% of AML blasts aberrantly express FLT3, making FLT3 an attractive therapeutic target for AML. Currently, several small molecule tyrosine kinase inhibitors (TKIs) targeting FLT3 have been approved in the treatment of AML, but they need to be used in combination with chemotherapy because of their limited potency to eliminate leukemic cells as single agents, largely due to the development of secondary inhibitor-resistant FLT3 mutations. Therefore, novel therapeutic strategies targeting FLT3 are needed. In the present study, we developed a FLT3 ligand-emtansine drug conjugate (FL-DM1) that targeted FLT3-positive AML cells with high potency and selectivity. We expressed recombinant human FLT3 ligand (rhFL) in the periplasm of recombinant E. Coli. The protein was purified by a two-step purification system containing Ni-NTA and Phenyl Sepharose. Our purified rhFL was bioactive to stimulate phosphorylation of FLT3 and proliferation of THP-1 cells. Next, we conjugated purified rhFL and emtansine (DM1) with SPDP linker. Reducing and non-reducing SDS-PAGE revealed that rhFL and DM1 were successfully conjugated, evidenced by a band with higher molecular weight of the conjugation product. Previous studies show that DM1 is a drug preferentially targeting proliferating cells by depolymerizing microtubules through binding at the vinca binding site of tubulin. We found that FL-DM1 reserved the physiological function of FLT3 ligand to stimulate proliferation of AML cells by inducing phosphorylation of FLT3 and the downstream signaling protein AKT in immunoblot, potentially enhancing the potency of FL-DM1 to inhibit FLT3-positive AML cells. Furthermore, flow cytometry showed that the surface expression of FLT3 significantly decreased on cells treated by FL-DM1 within two hours, which indicated the internalization of FL-DM1/FLT3 complex on these FLT3-positive AML cells, providing a mechanism of FL-DM1 entering target cells. In cell viability assay, we found that FL-DM1 effectively inhibited FLT3-positive AML cells THP-1 and MV-4-11 with IC50 around 10-30 nM. Also, FL-DM1 induced apoptosis and cell cycle arrest at G2/M phase in these cells detected by flow cytometry. In our previous studies, we generated FLT3-ITD transformed HCD-57 cells. HCD-57 cells are FLT3-negative erythroleukemia cells that depend on erythropoietin for survival. When infected with recombinant retroviruses carrying FLT3-ITD, they acquired ability to proliferate in the absence of EPO. We found that FL-DM1 inhibited HCD-57 cells transformed by FLT3-ITD, but not parental HCD-57 cells without FLT3 expression, indicating the selectivity of FL-DM1 to target FLT3-positive AML cells. In conclusion, our data demonstrated that FL-based drug conjugate can serve as an effective drug to target FLT3-expressing AML cells. Further studies will focus on in-vivo evaluation of FL-DM1 in animal models, the production of uncleavable SMCC linked FL-DM1 with improved in-vivo pharmacokinetic properties, and screening of FL muteins-DM1 conjugates with desired pharmacological properties. Disclosures No relevant conflicts of interest to declare.
- Published
- 2020
- Full Text
- View/download PDF
47. FLT3 Inhibitors Induce Instability of p53 By Mir-181 Mediated Downregulation of Deubiquitinase YOD1 in Acute Myeloid Leukemia
- Author
-
Han Zhong Pei, Yao Guo, Dengyang Zhang, Zhiguang Chang, Suk-Hwan Baek, Yun Chen, Liuting Yu, Bo Lu, Yuming Zhao, and Zhizhuang Joe Zhao
- Subjects
Myeloid ,biology ,Immunology ,Myeloid leukemia ,Cell Biology ,Hematology ,Biochemistry ,Transcriptome ,chemistry.chemical_compound ,medicine.anatomical_structure ,Downregulation and upregulation ,chemistry ,hemic and lymphatic diseases ,microRNA ,biology.protein ,medicine ,Cancer research ,Mdm2 ,Tyrosine kinase ,Quizartinib - Abstract
Acute myeloid leukemia (AML) is a hematological malignancy featured by impaired differentiation and uncontrolled proliferation of myeloid blasts. FLT3 internal tandem duplication (ITD) presents in 30-40% patients with AML, which serves as an independent poor prognostic marker and an attractive therapeutic target. Up to date, several tyrosine kinase inhibitors (TKIs) targeting FLT3 have been approved by FDA in the treatment of AML. However, FLT3 TKIs as single agents have limited effects to eliminate AML cells due to multiple drug-resistant mechanisms, including secondary FLT3 mutations, alternatively activated cell survival pathways, upregulation of FLT3 ligand, and downregulation of tumor suppressor genes. In the present study, we found that FLT3 TKIs decreased tumor suppressor p53 protein level by downregulation of YOD1 through miR-181 in FLT3-ITD mutant AML cells. In our previous studies, we generated FLT3-ITD transformed HCD-57 cells. HCD-57 cells are erythroleukemia cells that depend on erythropoietin for survival. When infected with recombinant retroviruses carrying FLT3-ITD, they acquired ability to proliferate in the absence of EPO. By using transcriptome analysis with RNAseq, we identified multiple differentially expressed miRNAs in HCD-57 transformed by FLT3-ITD, compared with parental HCD-57 cells. miR-181a-5p and miR-181b-5p were among these highly differentially expressed miRNAs. These two miRNAs were predicted to bind in 3'UTR of deubiquitinase YOD1 by using TargetScanHuman 7.2, an online tool to predict biological targets of miRNAs. Previous studies have shown that the ubiquitination and protein level of p53 is dysregulated due to overexpressed E3 ligase MDM2/4 in AML cells, but few studies focused on deubiquitinase of p53. We found that deubiquitinase YOD1 interacted with p53 by immunoprecipitation. Overexpression of YOD1 prevented degradation of p53 led by cycloheximide, a protein synthesis inhibitor. We further found that overexpression of YOD1 resulted in decreased ubiquitination of p53, indicating that YOD1 stabilized p53 protein through deubiquitination. Subsequently, we detected the expression of miR-181, YOD1 and p53 in a FLT3-ITD positive AML cell line MV-4-11 treated by FLT3 TKIs sorafenib, sunitinib and quizartinib. Real-time quantitative PCR showed that the treatment of FLT3 TKIs upregulated the expression of miR-181a-5p/miR-181b-5p, and downregulated mRNA level of their predicted target YOD1. The mRNA level of p53 remained unchanged but its protein level decreased with enhanced ubiquitination in MV-4-11 cells treated by FLT3 TKIs in the ubiquitination assay. These data suggested that FLT3 TKIs could reduce the stability of p53 by regulating miRNA-targeted YOD1. In addition, we collected peripheral blood mononuclear cells (PBMCs) from patients with AML and age-matched healthy donors. We found increased expression of miR-181a-5p and miR-181b-5p and decreased expression of YOD1 in PBMCs from AML patients compared with healthy donors, suggesting a pathological role of miR-181 to regulate YOD1/53 pathway in AML. In conclusion, our data showed that FLT3 TKIs induced instability of p53 by miR-181 mediated downregulation of YOD1. YOD1 as a novel deubiquitinase of p53 could play important roles in drug-resistance and progression of AML. Disclosures No relevant conflicts of interest to declare.
- Published
- 2020
- Full Text
- View/download PDF
48. CRISPR/Cas9 Knockout of Bak Mediates Bax Translocation to Mitochondria in response to TNF
- Author
-
Jingtian, Zhang, Han, Niu, Zhizhuang Joe, Zhao, Xueqi, Fu, Yuxiang, Wang, Xu, Zhang, Fuqiang, Zhang, and Linlin, Zeng
- Subjects
Tumor Necrosis Factor-alpha ,Apoptosis ,Mitochondria ,Gene Expression Regulation, Neoplastic ,Gene Knockout Techniques ,Cytosol ,bcl-2 Homologous Antagonist-Killer Protein ,Proto-Oncogene Proteins c-bcl-2 ,Caspases ,Cell Line, Tumor ,Humans ,biological phenomena, cell phenomena, and immunity ,CRISPR-Cas Systems ,Cycloheximide ,RNA, Small Interfering ,bcl-2-Associated X Protein ,Research Article - Abstract
TNFα/CHX-induced apoptosis is dependent on caspase-8 activation and regulated by Bcl-2. However, the specific participants and precise mechanisms underlying this apoptotic pathway are poorly understood. The proapoptotic proteins Bak and Bax—members of the Bcl-2 family—are essential for the functioning of the mitochondrial apoptotic pathway. In this study, we used the CRISPR/Cas9 system to knockout Bak in the human SH-SY5Y cell line and determined the effects of this knockout on TNFα/CHX-induced apoptosis. Our data showed that overexpression of Bcl-2 dramatically prevented TNFα/CHX-induced apoptosis, and then pro-apoptotic protein Bak was downregulated and became more resistant to TNFα/CHX-induced apoptosis, because both TNFα/CHX-induced PARP cleavage and caspase activation were blocked in BAK−/− cells or using specific siRNA, whereas Bax was dispensable in TNFα/CHX-induced apoptosis, as evidenced using specific siRNA. Bax translocated from the cytosol into the mitochondria in response to TNFα/CHX, and CRISPR/Cas9 knockout of Bak significantly decreased this translocation. These results indicate that TNFα/CHX-induced apoptosis does not occur in Bak−/− cells, suggesting that TNFα/CHX-induced apoptosis is Bak-dependent but Bax-independent.
- Published
- 2019
49. CRISPR/Cas9 Knockout of Bak Mediates Bax Translocation to Mitochondria in response to TNFα/CHX-induced Apoptosis
- Author
-
Xueqi Fu, Wang Yuxiang, Fuqiang Zhang, Linlin Zeng, Zhizhuang Joe Zhao, Jingtian Zhang, Xu Zhang, and Han Niu
- Subjects
0301 basic medicine ,Regulation of gene expression ,General Immunology and Microbiology ,Article Subject ,Chemistry ,lcsh:R ,lcsh:Medicine ,Chromosomal translocation ,General Medicine ,Mitochondrion ,General Biochemistry, Genetics and Molecular Biology ,Cell biology ,03 medical and health sciences ,Cytosol ,030104 developmental biology ,0302 clinical medicine ,Apoptosis ,Cell culture ,030220 oncology & carcinogenesis ,Gene Knockout Techniques ,Tumor necrosis factor alpha ,biological phenomena, cell phenomena, and immunity - Abstract
TNFα/CHX-induced apoptosis is dependent on caspase-8 activation and regulated by Bcl-2. However, the specific participants and precise mechanisms underlying this apoptotic pathway are poorly understood. The proapoptotic proteins Bak and Bax—members of the Bcl-2 family—are essential for the functioning of the mitochondrial apoptotic pathway. In this study, we used the CRISPR/Cas9 system to knockout Bak in the human SH-SY5Y cell line and determined the effects of this knockout on TNFα/CHX-induced apoptosis. Our data showed that overexpression of Bcl-2 dramatically prevented TNFα/CHX-induced apoptosis, and then pro-apoptotic protein Bak was downregulated and became more resistant to TNFα/CHX-induced apoptosis, because both TNFα/CHX-induced PARP cleavage and caspase activation were blocked in BAK−/− cells or using specific siRNA, whereas Bax was dispensable in TNFα/CHX-induced apoptosis, as evidenced using specific siRNA. Bax translocated from the cytosol into the mitochondria in response to TNFα/CHX, and CRISPR/Cas9 knockout of Bak significantly decreased this translocation. These results indicate that TNFα/CHX-induced apoptosis does not occur in Bak−/− cells, suggesting that TNFα/CHX-induced apoptosis is Bak-dependent but Bax-independent.
- Published
- 2019
- Full Text
- View/download PDF
50. ASXL1 alteration cooperates with JAK2V617F to accelerate myelofibrosis
- Author
-
Ying, Guo, Yuan, Zhou, Shohei, Yamatomo, Hui, Yang, Peng, Zhang, Shi, Chen, Stephen D, Nimer, Zhizhuang Joe, Zhao, Mingjiang, Xu, Jie, Bai, and Feng-Chun, Yang
- Subjects
Repressor Proteins ,Amino Acid Substitution ,Gene Expression Regulation ,Primary Myelofibrosis ,Mutation ,Disease Progression ,Humans ,Epistasis, Genetic ,Genetic Predisposition to Disease ,Janus Kinase 2 ,Prognosis - Published
- 2018
Catalog
Discovery Service for Jio Institute Digital Library
For full access to our library's resources, please sign in.