Network


Latest external collaboration on country level. Dive into details by clicking on the dots.

Hotspot


Dive into the research topics where Zhihong Fang is active.

Publication


Featured researches published by Zhihong Fang.


Leukemia | 2014

Expression of PD-L1, PD-L2, PD-1 and CTLA4 in myelodysplastic syndromes is enhanced by treatment with hypomethylating agents

Hui Yang; Carlos E. Bueso-Ramos; Courtney D. DiNardo; Marcos R. Estecio; Masoud Davanlou; Qirong Geng; Zhihong Fang; Martin Nguyen; Sherry Pierce; Yue Wei; Simrit Parmar; Jorge Cortes; Hagop M. Kantarjian; Guillermo Garcia-Manero

Blockade of immune checkpoints is emerging as a new form of anticancer therapy. We studied the expression of programmed death ligand 1 (PD-L1), PD-L2, programmed death 1 (PD-1) and cytotoxic T lymphocyte-associated antigen 4 (CTLA4) mRNA in CD34+ cells from myelodysplastic syndrome (MDS), chronic myelomonocytic leukemia (CMML) and acute myeloid leukemia (AML) patients (N=124). Aberrant upregulation (⩾2-fold) was observed in 34, 14, 15 and 8% of the patients. Increased expression of these four genes was also observed in peripheral blood mononuclear cells (PBMNCs) (N=61). The relative expression of PD-L1 from PBMNC was significantly higher in MDS (P=0.018) and CMML (P=0.0128) compared with AML. By immunohistochemical analysis, PD-L1 protein expression was observed in MDS CD34+ cells, whereas stroma/non-blast cellular compartment was positive for PD-1. In a cohort of patients treated with epigenetic therapy, PD-L1, PD-L2, PD-1 and CTLA4 expression was upregulated. Patients resistant to therapy had relative higher increments in gene expression compared with patients who achieved response. Treatment of leukemia cells with decitabine resulted in a dose-dependent upregulation of above genes. Exposure to decitabine resulted in partial demethylation of PD-1 in leukemia cell lines and human samples. This study suggests that PD-1 signaling may be involved in MDS pathogenesis and resistance mechanisms to hypomethylating agents. Blockade of this pathway can be a potential therapy in MDS and AML.


Leukemia | 2008

Genome-wide identification of aberrantly methylated promoter associated CpG islands in acute lymphocytic leukemia

Shaoqing Kuang; Weigang Tong; Hui Yang; W. Lin; Ming Lee; Zhihong Fang; Yue Wei; Jaroslav Jelinek; J. P. Issa; Guillermo Garcia-Manero

We performed a genome-wide analysis of promoter associated CpG island methylation using methylated CpG island amplification (MCA) coupled to representational differential analysis (RDA) or a DNA promoter microarray in acute lymphoblastic leukemia (ALL). We identified 65 potential targets of methylation with the MCA/RDA approach, and 404 with the MCA/array. Thirty-six (77%) of the genes identified by MCA/RDA were shared by the MCA/array approach. Chromosomal location of these genes was evenly distributed in all autosomes. Functionally, 303 of these genes clustered in 18 molecular pathways. Of the 36 shared genes, 31 were validated and 26 were confirmed as being hypermethylated in leukemia cell lines. Expression analysis of eight of these genes was epigenetically modulated by hypomethylating agents and/or HDAC inhibitors in leukemia cell lines. Subsequently, DNA methylation of 15 of these genes (GIPC2, RSPO1, MAGI1, CAST1, ADCY5, HSPA4L, OCLN, EFNA5, MSX2, GFPT2, GNA14, SALL1, MYO5B, ZNF382 and MN1) was validated in primary ALL samples. Patients with methylation of multiple CpG islands had a worse overall survival. This is the largest published list of potential methylation target genes in human leukemia offering the possibility of performing rational unbiased methylation studies in ALL.


Leukemia | 2013

Global H3K4me3 genome mapping reveals alterations of innate immunity signaling and overexpression of JMJD3 in human myelodysplastic syndrome CD34+ cells

Yue Wei; Rui Chen; Sophie Dimicoli; Carlos E. Bueso-Ramos; Donna Neuberg; Sherry Pierce; Hui Wang; Hui Yang; Yu Jia; Hong Zheng; Zhihong Fang; Martin Nguyen; Irene Gañán-Gómez; Benjamin L. Ebert; Ross L. Levine; Hagop M. Kantarjian; Guillermo Garcia-Manero

The molecular bases of myelodysplastic syndromes (MDS) are not fully understood. Trimethylated histone 3 lysine 4 (H3K4me3) is present in promoters of actively transcribed genes and has been shown to be involved in hematopoietic differentiation. We performed a genome-wide H3K4me3 CHIP-Seq (chromatin immunoprecipitation coupled with whole genome sequencing) analysis of primary MDS bone marrow (BM) CD34+ cells. This resulted in the identification of 36 genes marked by distinct higher levels of promoter H3K4me3 in MDS. A majority of these genes are involved in nuclear factor (NF)-κB activation and innate immunity signaling. We then analyzed expression of histone demethylases and observed significant overexpression of the JmjC-domain histone demethylase JMJD3 (KDM6b) in MDS CD34+ cells. Furthermore, we demonstrate that JMJD3 has a positive effect on transcription of multiple CHIP-Seq identified genes involved in NF-κB activation. Inhibition of JMJD3 using shRNA in primary BM MDS CD34+ cells resulted in an increased number of erythroid colonies in samples isolated from patients with lower-risk MDS. Taken together, these data indicate the deregulation of H3K4me3 and associated abnormal activation of innate immunity signals have a role in the pathogenesis of MDS and that targeting these signals may have potential therapeutic value in MDS.


PLOS ONE | 2013

Overexpression of the Toll-Like Receptor (TLR) Signaling Adaptor MYD88, but Lack of Genetic Mutation, in Myelodysplastic Syndromes

Sophie Dimicoli; Yue Wei; Carlos E. Bueso-Ramos; Hui Yang; Courtney D. DiNardo; Yu Jia; Hong Zheng; Zhihong Fang; Martin Nguyen; Sherry Pierce; Rui Chen; Hui Wang; Chenghua Wu; Guillermo Garcia-Manero

MYD88 is a key mediator of Toll-like receptor innate immunity signaling. Oncogenically active MYD88 mutations have recently been reported in lymphoid malignancies, but has not been described in MDS. To characterize MYD88 in MDS, we sequenced the coding region of the MYD88 gene in 40 MDS patients. No MYD88 mutation was detected. We next characterized MYD88 expression in bone marrow CD34+ cells (N = 64). Increased MYD88 RNA was detected in 40% of patients. Patients with higher MYD88 expression in CD34+ cells had a tendency for shorter survival compared to the ones with lower MYD88, which was significant when controlled for IPSS and age. We then evaluated effect of MYD88 blockade in the CD34+ cells of patients with lower-risk MDS. Colony formation assays indicated that MYD88 blockade using a MYD88 inhibitor resulted in increased erythroid colony formation. MYD88 blockade also negatively regulated the secretion of interleukin-8. Treatment of MDS CD34+ cells with an IL-8 antibody also increased formation of erythroid colonies. These results indicate that MYD88 plays a role in the pathobiology of MDS and may have prognostic and therapeutic value in the management of patients with this disease.


PLOS ONE | 2013

Epigenetic Inactivation of Notch-Hes Pathway in Human B-Cell Acute Lymphoblastic Leukemia

Shao Qing Kuang; Zhihong Fang; Patrick A. Zweidler-McKay; Hui Yang; Yue Wei; Emilio A. Gonzalez-Cervantes; Yanis Boumber; Guillermo Garcia-Manero

The Notch pathway can have both oncogenic and tumor suppressor roles, depending on cell context. For example, Notch signaling promotes T cell differentiation and is leukemogenic in T cells, whereas it inhibits early B cell differentiation and acts as a tumor suppressor in B cell leukemia where it induces growth arrest and apoptosis. The regulatory mechanisms that contribute to these opposing roles are not understood. Aberrant promoter DNA methylation and histone modifications are associated with silencing of tumor suppressor genes and have been implicated in leukemogenesis. Using methylated CpG island amplification (MCA)/DNA promoter microarray, we identified Notch3 and Hes5 as hypermethylated in human B cell acute lymphoblastic leukemia (ALL). We investigated the methylation status of other Notch pathway genes by bisulfite pyrosequencing. Notch3, JAG1, Hes2, Hes4 and Hes5 were frequently hypermethylated in B leukemia cell lines and primary B-ALL, in contrast to T-ALL cell lines and patient samples. Aberrant methylation of Notch3 and Hes5 in B-ALL was associated with gene silencing and was accompanied by decrease of H3K4 trimethylation and H3K9 acetylation and gain of H3K9 trimethylation and H3K27 trimethylation. 5-aza-2′-deoxycytidine treatment restored Hes5 expression and decreased promoter hypermethylation in most leukemia cell lines and primary B-ALL samples. Restoration of Hes5 expression by lentiviral transduction resulted in growth arrest and apoptosis in Hes5 negative B-ALL cells but not in Hes5 expressing T-ALL cells. These data suggest that epigenetic modifications are implicated in silencing of tumor suppressor of Notch/Hes pathway in B-ALL.


Leukemia Research | 2010

Preclinical antileukemia activity of JNJ-26481585, a potent second-generation histone deacetylase inhibitor.

Wei Gang Tong; Yue Wei; William Stevenson; Shao Qing Kuang; Zhihong Fang; Ming Zhang; Janine Arts; Guillermo Garcia-Manero

Histone deacetylase (HDAC) inhibitors have been shown to induce cell cycle arrest, terminal differentiation, and apoptosis in a broad spectrum of human tumors and animal xenograft models. JNJ-26481585 is a hydroxamic acid derivative, second-generation pan-HDAC inhibitor that has demonstrated high potency in preclinical studies. In the current study, we demonstrated that JNJ-26481585 has antileukemia and molecular activity in leukemia cell lines and primary human leukemia cells. We also observed a synergistic effect between treatment with decitabine and JNJ-26481585. In conclusion, JNJ-26481585 is a potent second-generation pan-HDAC inhibitor with activity in human leukemia, and it is currently in clinical development.


British Journal of Haematology | 2015

RUNX3 promoter hypermethylation is frequent in leukaemia cell lines and associated with acute myeloid leukaemia inv(16) subtype

Marcos R. Estecio; Sirisha Maddipoti; Carlos E. Bueso-Ramos; Courtney D. DiNardo; Hui Yang; Yue Wei; Kimie Kondo; Zhihong Fang; William Stevenson; Kun Sang Chang; Sherry Pierce; Zachary S. Bohannan; Gautam Borthakur; Hagop M. Kantarjian; Guillermo Garcia-Manero

Correlative and functional studies support the involvement of the RUNX gene family in haematological malignancies. To elucidate the role of epigenetics in RUNX inactivation, we evaluated promoter DNA methylation of RUNX1, 2, and 3 in 23 leukaemia cell lines and samples from acute myeloid leukaemia (AML), acute lymphocytic leukaemia (ALL) and myelodysplatic syndromes (MDS) patients. RUNX1 and RUNX2 gene promoters were mostly unmethylated in cell lines and clinical samples. Hypermethylation of RUNX3 was frequent among cell lines (74%) and highly variable among patient samples, with clear association to cytogenetic status. High frequency of RUNX3 hypermethylation (85% of the 20 studied cases) was found in AML patients with inv(16)(p13.1q22) compared to other AML subtypes (31% of the other 49 cases). RUNX3 hypermethylation was also frequent in ALL (100% of the six cases) but low in MDS (21%). In support of a functional role, hypermethylation of RUNX3 was correlated with low levels of protein, and treatment of cell lines with the DNA demethylating agent, decitabine, resulted in mRNA re‐expression. Furthermore, relapse‐free survival of non‐inv(16)(p13.1q22) AML patients without RUNX3 methylation was significantly better (P = 0·016) than that of methylated cases. These results suggest that RUNX3 silencing is an important event in inv(16)(p13.1q22) leukaemias.


Leukemia Research | 2017

Preclinical activity of FF-10501-01, a novel inosine-5′-monophosphate dehydrogenase inhibitor, in acute myeloid leukemia

Hui Yang; Zhihong Fang; Yue Wei; Zachary S. Bohannan; Irene Ganan-Gomez; Ana Alfonso Pierola; Hiroyuki Iwamura; Guillermo Garcia-Manero

BACKGROUND FF-10501-01 is a selective inosine monophosphate dehydrogenase (IMPDH) inhibitor that has shown activity in cancer cell lines. We studied whether FF-10501-01 is effective in targeting a variety of hypomethylating agent (HMA)-sensitive and -resistant acute myelogenous leukemia (AML) cell lines. METHODS We treated multiple cell lines (including HMA-resistant cells) with FF-10501-01 and analyzed proliferation, apoptosis, and cell cycle status. We also assessed HMA-FF-10501-01 combinations and the ability of extracellular guanosine to rescue cell proliferation in FF-10501-01-treated cells. We performed high-performance liquid chromatography (HPLC) to study guanine nucleotide levels in treated and untreated cells. Finally, we studied the effects of FF-10501-01 in fresh peripheral blood cells taken from AML patients. RESULTS FF-10501-01 showed a strong dose-dependent effect on proliferation and induced apoptosis at approximately 30μM. The effects of FF-10501-01 treatment on cell cycle status were variable, with no statistically significant trends. Guanosine rescued proliferation in FF-10501-01-treated cells, and HPLC results showed significant decreases in phosphorylated guanosine levels in MOLM13 cells. FF-10501-01 effectively reduced proliferation at concentrations of 300μM and above in 3 primary AML samples. CONCLUSIONS FF-10501-01 effectively induces AML cell death and reduces AML peripheral blood cell proliferation by targeting guanine nucleotide biosynthesis regardless of HMA resistance status.


Clinical Lymphoma, Myeloma & Leukemia | 2015

Underexpression of EZH2 in Myelodysplastic Syndromes

Yue Wei; Monica Cabrero; Yu Jia; Hong Zheng; Hui Yang; Zhihong Fang; Zach Bohannan; Guillermo Garcia-Manero

Results: We found that 47% (n=37) of our samples have reduced EZH2 mRNA expression (less than half that of controls), but these data not statistically significant (p=0.159). Further analyses revealed that EZH2 is significantly underexpressed in MDS cases with chromosome 7 or 7q deletion (mean=0.4 fold, p=0.006). These deletions were also associated with lower EZH2 expression than was seen in diploid cells and cells with other cytogenetic abnormalities (p=0.041). To understand the effects of EZH2 expression alteration specifically, we performed survival analysis. Multivariate analysis indicated that, in contrast to previous findings, EZH2 underexpression is associated with better overall survival (OS) (p=0.012). We also examined treatments in our patient cohort and found that 61% of patients (n=53) received hypomethylating agents (HMA). In the HMA treatment cohort, non-responding patients (n=27) had lower EZH2 expression than responders (n=26), but that effect was not significant (p=0.12). However, in HMA responders, EZH2 expression was significantly lower in patients who achieved longer responses (more than 12 months, n=15) than in those who progressed or relapsed within 12 months (p=0.02).


American Journal of Hematology | 2011

Levels of miR-29b do not predict for response in patients with acute myelogenous leukemia treated with the combination of 5-azacytidine, valproic acid, and ATRA

Hui Yang; Zhihong Fang; Yue Wei; Yumin Hu; George A. Calin; Hagop M. Kantarjian; Guillermo Garcia-Manero

Collaboration


Dive into the Zhihong Fang's collaboration.

Top Co-Authors

Avatar

Guillermo Garcia-Manero

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Yue Wei

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hagop M. Kantarjian

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Hong Zheng

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Sherry Pierce

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Carlos E. Bueso-Ramos

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Yu Jia

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Martin Nguyen

University of Texas MD Anderson Cancer Center

View shared research outputs
Top Co-Authors

Avatar

Courtney D. DiNardo

University of Texas MD Anderson Cancer Center

View shared research outputs
Researchain Logo
Decentralizing Knowledge