Network


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

Hotspot


Dive into the research topics where Yanming Zhang is active.

Publication


Featured researches published by Yanming Zhang.


Proceedings of the National Academy of Sciences of the United States of America | 2007

MicroRNA expression signatures accurately discriminate acute lymphoblastic leukemia from acute myeloid leukemia

Shuangli Mi; Jun Lu; Miao Sun; Zejuan Li; Hao Zhang; Mary Beth Neilly; Yungui Wang; Zhijian Qian; Jie Jin; Yanming Zhang; Stefan K. Bohlander; Michelle M. Le Beau; Richard A. Larson; Todd R. Golub; Janet D. Rowley; Jianjun Chen

Acute lymphoblastic leukemia (ALL) is the most common childhood cancer, whereas acute myeloid leukemia (AML) is the most common acute leukemia in adults. In general, ALL has a better prognosis than AML. To understand the distinct mechanisms in leukemogenesis between ALL and AML and to identify markers for diagnosis and treatment, we performed a large-scale genome-wide microRNA (miRNA, miR) expression profiling assay and identified 27 miRNAs that are differentially expressed between ALL and AML. Among them, miR-128a and -128b are significantly overexpressed, whereas let-7b and miR-223 are significantly down-regulated in ALL compared with AML. They are the most discriminatory miRNAs between ALL and AML. Using the expression signatures of a minimum of two of these miRNAs resulted in an accuracy rate of >95% in the diagnosis of ALL and AML. The differential expression patterns of these four miRNAs were validated further through large-scale real-time PCR on 98 acute leukemia samples covering most of the common cytogenetic subtypes, along with 10 normal control samples. Furthermore, we found that overexpression of miR-128 in ALL was at least partly associated with promoter hypomethylation and not with an amplification of its genomic locus. Taken together, we showed that expression signatures of as few as two miRNAs could accurately discriminate ALL from AML, and that epigenetic regulation might play an important role in the regulation of expression of miRNAs in acute leukemias.


Proceedings of the National Academy of Sciences of the United States of America | 2008

Distinct microRNA expression profiles in acute myeloid leukemia with common translocations

Zejuan Li; Jun Lu; Miao Sun; Shuangli Mi; Hao Zhang; Roger T. Luo; Ping Chen; Yungui Wang; Ming Yan; Zhijian Qian; Mary Beth Neilly; Jie Jin; Yanming Zhang; Stefan K. Bohlander; Dong-Er Zhang; Richard A. Larson; Michelle M. Le Beau; Michael J. Thirman; Todd R. Golub; Janet D. Rowley; Jianjun Chen

MicroRNAs (miRNAs) are postulated to be important regulators in cancers. Here, we report a genome-wide miRNA expression analysis in 52 acute myeloid leukemia (AML) samples with common translocations, including t(8;21)/AML1(RUNX1)-ETO(RUNX1T1), inv(16)/CBFB-MYH11, t(15;17)/PML-RARA, and MLL rearrangements. Distinct miRNA expression patterns were observed for t(15;17), MLL rearrangements, and core-binding factor (CBF) AMLs including both t(8;21) and inv(16) samples. Expression signatures of a minimum of two (i.e., miR-126/126*), three (i.e., miR-224, miR-368, and miR-382), and seven (miR-17–5p and miR-20a, plus the aforementioned five) miRNAs could accurately discriminate CBF, t(15;17), and MLL-rearrangement AMLs, respectively, from each other. We further showed that the elevated expression of miR-126/126* in CBF AMLs was associated with promoter demethylation but not with amplification or mutation of the genomic locus. Our gain- and loss-of-function experiments showed that miR-126/126* inhibited apoptosis and increased the viability of AML cells and enhanced the colony-forming ability of mouse normal bone marrow progenitor cells alone and particularly, in cooperation with AML1-ETO, likely through targeting Polo-like kinase 2 (PLK2), a tumor suppressor. Our results demonstrate that specific alterations in miRNA expression distinguish AMLs with common translocations and imply that the deregulation of specific miRNAs may play a role in the development of leukemia with these associated genetic rearrangements.


Journal of Clinical Oncology | 2009

Pomalidomide is active in the treatment of anemia associated with myelofibrosis

Ayalew Tefferi; Srdan Verstovsek; Giovanni Barosi; Francesco Passamonti; Gail J. Roboz; Heinz Gisslinger; Ronald Paquette; Francisco Cervantes; Candido E. Rivera; H. Joachim Deeg; Juergen Thiele; Hans Michael Kvasnicka; James W. Vardiman; Yanming Zhang; B. Nebiyou Bekele; Ruben A. Mesa; Robert Peter Gale; Hagop M. Kantarjian

PURPOSE Thalidomide and lenalidomide can alleviate anemia in myelofibrosis. However, their value is undermined by their respective potential to cause peripheral neuropathy and myelosuppression. We therefore evaluated the safety and therapeutic activity of another immunomodulatory drug, pomalidomide. METHODS In a phase II randomized, multicenter, double-blind, adaptive design study, four treatment arms were evaluated: pomalidomide (2 mg/d) plus placebo, pomalidomide (2 mg/d) plus prednisone, pomalidomide (0.5 mg/d) plus prednisone, and prednisone plus placebo. Pomalidomide was administered for up to 12 28-day treatment cycles. Prednisone (30 mg/d) was given in a tapering dose schedule during the first three cycles. Response was assessed by International Working Group criteria. RESULTS Eighty-four patients with myelofibrosis-associated anemia were randomly assigned to the aforementioned treatment arms: 22, 19, 22, and 21, respectively. Response in anemia was documented in 20 patients, including 15 who became transfusion independent. Response rates in the four treatment arms were 23% (95% CI, 5% to 41%), 16% (95% CI, 0% to 33%), 36% (95% CI, 16% to 56%), and 19% (95% CI, 2% to 36%). The corresponding figures for patients receiving > or = 3 cycles of treatment (n = 62) were 38%, 23%, 40%, and 25%. Response to pomalidomide with or without prednisone was durable (range, 3.2 to 16.9+ months) and significantly better in the absence of leukocytosis (37% v 8%; P = .01); JAK2V617F or cytogenetic status did not affect response. Grade > or = 3 toxicities were infrequent and included (in each treatment arm) neutropenia (9%; 16%; 5%; 5%), thrombocytopenia (14%; 16%; 9%; 5%), and thrombosis (9%; 5%; 0%; 0%). CONCLUSION Pomalidomide therapy at 0.5 or 2 mg/d with or without an abbreviated course of prednisone is well tolerated in patients with myelofibrosis and active in the treatment of anemia.


Blood | 2012

Up-regulation of a HOXA-PBX3 homeobox-gene signature following down-regulation of miR-181 is associated with adverse prognosis in patients with cytogenetically abnormal AML

Zejuan Li; Hao Huang; Yuanyuan Li; Xi Jiang; Ping Chen; Stephen Arnovitz; Michael D. Radmacher; Kati Maharry; Abdel G. Elkahloun; Xinan Yang; Chunjiang He; Miao He; Zhiyu Zhang; Konstanze Döhner; Mary Beth Neilly; Colles Price; Yves A. Lussier; Yanming Zhang; Richard A. Larson; Michelle M. Le Beau; Michael A. Caligiuri; Lars Bullinger; Ruud Delwel; Bob Löwenberg; Paul Liu; Guido Marcucci; Clara D. Bloomfield; Janet D. Rowley; Jianjun Chen

Increased expression levels of miR-181 family members have been shown to be associated with favorable outcome in patients with cytogenetically normal acute myeloid leukemia. Here we show that increased expression of miR-181a and miR-181b is also significantly (P < .05; Cox regression) associated with favorable overall survival in cytogenetically abnormal AML (CA-AML) patients. We further show that up-regulation of a gene signature composed of 4 potential miR-181 targets (including HOXA7, HOXA9, HOXA11, and PBX3), associated with down-regulation of miR-181 family members, is an independent predictor of adverse overall survival on multivariable testing in analysis of 183 CA-AML patients. The independent prognostic impact of this 4-homeobox-gene signature was confirmed in a validation set of 271 CA-AML patients. Furthermore, our in vitro and in vivo studies indicated that ectopic expression of miR-181b significantly promoted apoptosis and inhibited viability/proliferation of leukemic cells and delayed leukemogenesis; such effects could be reversed by forced expression of PBX3. Thus, the up-regulation of the 4 homeobox genes resulting from the down-regulation of miR-181 family members probably contribute to the poor prognosis of patients with nonfavorable CA-AML. Restoring expression of miR-181b and/or targeting the HOXA/PBX3 pathways may provide new strategies to improve survival substantially.


Proceedings of the National Academy of Sciences of the United States of America | 2002

Genomic DNA breakpoints in AML1/RUNX1 and ETO cluster with topoisomerase II DNA cleavage and DNase I hypersensitive sites in t(8;21) leukemia

Yanming Zhang; Pamela L. Strissel; Reiner Strick; Jianjun Chen; Giuseppina Nucifora; Michelle M. Le Beau; Richard A. Larson; Janet D. Rowley

The translocation t(8;21)(q22;q22) is one of the most frequent chromosome translocations in acute myeloid leukemia (AML). AML1/RUNX1 at 21q22 is involved in t(8;21), t(3;21), and t(16;21) in de novo and therapy-related AML and myelodysplastic syndrome as well as in t(12;21) in childhood B cell acute lymphoblastic leukemia. Although DNA breakpoints in AML1 and ETO (at 8q22) cluster in a few introns, the mechanisms of DNA recombination resulting in t(8;21) are unknown. The correlation of specific chromatin structural elements, i.e., topoisomerase II (topo II) DNA cleavage sites, DNase I hypersensitive sites, and scaffold-associated regions, which have been implicated in chromosome recombination with genomic DNA breakpoints in AML1 and ETO in t(8;21) is unknown. The breakpoints in AML1 and ETO were clustered in the Kasumi 1 cell line and in 31 leukemia patients with t(8;21); all except one had de novo AML. Sequencing of the breakpoint junctions revealed no common DNA motif; however, deletions, duplications, microhomologies, and nontemplate DNA were found. Ten in vivo topo II DNA cleavage sites were mapped in AML1, including three in intron 5 and seven in intron 7a, and two were in intron 1b of ETO. All strong topo II sites colocalized with DNase I hypersensitive sites and thus represent open chromatin regions. These sites correlated with genomic DNA breakpoints in both AML1 and ETO, thus implicating them in the de novo 8;21 translocation.


Archive | 1999

Classical and Molecular Cytogenetics of Tumor Cells

Brigitte Schlegelberger; Simone Metzke; Svetlana Harder; Reina Zühlke-Jenisch; Yanming Zhang; Reiner Siebert

Cytogenetic findings are becoming increasingly important for the management of patients with malignant diseases, especially for those with hematologic neoplasias. The detection of aquired somatic mutations may help to establish the diagnosis of a neoplastic disorder and to rule out reactive changes due to toxic injury, vitamin deficiency or infections. Before, however, a chromosome aberration found in tumor cells can be taken as tumorassociated change it should be ruled out by chromosome analysis on PHA-stimulated blood lymphocytes that this chromosome aberration does not represent a constitutional abnormality. It is now clear that certain so-called primary chromosome abnormalities of tumor cells are associated with distinct clinico-histological disease entities. During tumor evolution additional chromosome aberrations appear and may determine the clinical course of the disease. Even these so-called secondary chromosome aberrations are non-randomly distributed throughout the genome. Therefore, cytogenetic studies are essential to make a specified diagnosis, to classify malignant disorders, to characterize the degree of neoplastic progression, to predict the prognosis, to test for remission, and to establish when relapse occurs. Thus, cytogenetic data can be of great help to select the appropriate treatment strategy.


Journal of Clinical Oncology | 2013

Identification of a 24-Gene Prognostic Signature That Improves the European LeukemiaNet Risk Classification of Acute Myeloid Leukemia: An International Collaborative Study

Zejuan Li; Tobias Herold; Chunjiang He; Ping Chen; Vindi Jurinovic; Ulrich Mansmann; Michael D. Radmacher; Kati Maharry; Miao Sun; Xinan Yang; Hao Huang; Xi Jiang; Maria Cristina Sauerland; Thomas Büchner; Wolfgang Hiddemann; Abdel G. Elkahloun; Mary Beth Neilly; Yanming Zhang; Richard A. Larson; Michelle M. Le Beau; Michael A. Caligiuri; Konstanze Döhner; Lars Bullinger; Paul Liu; Ruud Delwel; Guido Marcucci; Bob Löwenberg; Clara D. Bloomfield; Janet D. Rowley; Stefan K. Bohlander

PURPOSE To identify a robust prognostic gene expression signature as an independent predictor of survival of patients with acute myeloid leukemia (AML) and use it to improve established risk classification. PATIENTS AND METHODS Four independent sets totaling 499 patients with AML carrying various cytogenetic and molecular abnormalities were used as training sets. Two independent patient sets composed of 825 patients were used as validation sets. Notably, patients from different sets were treated with different protocols, and their gene expression profiles were derived using different microarray platforms. Cox regression and Kaplan-Meier methods were used for survival analyses. RESULTS A prognostic signature composed of 24 genes was derived from a meta-analysis of Cox regression values of each gene across the four training sets. In multivariable models, a higher sum value of the 24-gene signature was an independent predictor of shorter overall (OS) and event-free survival (EFS) in both training and validation sets (P < .01). Moreover, this signature could substantially improve the European LeukemiaNet (ELN) risk classification of AML, and patients in three new risk groups classified by the integrated risk classification showed significantly (P < .001) distinct OS and EFS. CONCLUSION Despite different treatment protocols applied to patients and use of different microarray platforms for expression profiling, a common prognostic gene signature was identified as an independent predictor of survival of patients with AML. The integrated risk classification incorporating this gene signature provides a better framework for risk stratification and outcome prediction than the ELN classification.


Cancer Research | 2009

Consistent deregulation of gene expression between human and murine MLL rearrangement leukemias.

Zejuan Li; Roger T. Luo; Shuangli Mi; Miao Sun; Ping Chen; Jingyue Bao; Mary Beth Neilly; Nimanthi Jayathilaka; Deborah S. Johnson; Lili Wang; Catherine Lavau; Yanming Zhang; Charles C. Tseng; Xiuqing Zhang; Jian Wang; Jun Yu; Huanming Yang; San Ming Wang; Janet D. Rowley; Jianjun Chen; Michael J. Thirman

Important biological and pathologic properties are often conserved across species. Although several mouse leukemia models have been well established, the genes deregulated in both human and murine leukemia cells have not been studied systematically. We performed a serial analysis of gene expression in both human and murine MLL-ELL or MLL-ENL leukemia cells and identified 88 genes that seemed to be significantly deregulated in both types of leukemia cells, including 57 genes not reported previously as being deregulated in MLL-associated leukemias. These changes were validated by quantitative PCR. The most up-regulated genes include several HOX genes (e.g., HOX A5, HOXA9, and HOXA10) and MEIS1, which are the typical hallmark of MLL rearrangement leukemia. The most down-regulated genes include LTF, LCN2, MMP9, S100A8, S100A9, PADI4, TGFBI, and CYBB. Notably, the up-regulated genes are enriched in gene ontology terms, such as gene expression and transcription, whereas the down-regulated genes are enriched in signal transduction and apoptosis. We showed that the CpG islands of the down-regulated genes are hypermethylated. We also showed that seven individual microRNAs (miRNA) from the mir-17-92 cluster, which are overexpressed in human MLL rearrangement leukemias, are also consistently overexpressed in mouse MLL rearrangement leukemia cells. Nineteen possible targets of these miRNAs were identified, and two of them (i.e., APP and RASSF2) were confirmed further by luciferase reporter and mutagenesis assays. The identification and validation of consistent changes of gene expression in human and murine MLL rearrangement leukemias provide important insights into the genetic base for MLL-associated leukemogenesis.


Proceedings of the National Academy of Sciences of the United States of America | 2012

miR-495 is a tumor-suppressor microRNA down-regulated in MLL-rearranged leukemia

Xi Jiang; Hao Huang; Zejuan Li; Chunjiang He; Yuanyuan Li; Ping Chen; Sandeep Gurbuxani; Stephen Arnovitz; Gia Ming Hong; Colles Price; Haomin Ren; Rejani B. Kunjamma; Mary Beth Neilly; Justin Salat; Mark Wunderlich; Robert K. Slany; Yanming Zhang; Richard A. Larson; Michelle M. Le Beau; James C. Mulloy; Janet D. Rowley; Jianjun Chen

Acute myeloid leukemia (AML) is a heterogeneous group of hematopoietic malignancies with variable response to treatment. AMLs bearing MLL (mixed lineage leukemia) rearrangements are associated with intermediate or poor survival. MicroRNAs (miRNAs), a class of small noncoding RNAs, have been postulated to be important gene expression regulators virtually in all biological processes, including leukemogenesis. Through a large-scale, genome-wide miRNA expression profiling assay of 85 human AML and 15 normal control samples, we show that among 48 miRNAs that are significantly differentially expressed between MLL- and non–MLL-rearranged AML samples, only one (miR-495) is expressed at a lower level in MLL-rearranged AML than in non–MLL-rearranged AML; meanwhile, miR-495 is also significantly down-regulated in MLL-rearranged AML samples compared with normal control samples. Through in vitro colony-forming/replating assays and in vivo bone marrow transplantation studies, we show that forced expression of miR-495 significantly inhibits MLL-fusion-mediated cell transformation in vitro and leukemogenesis in vivo. In human leukemic cells carrying MLL rearrangements, ectopic expression of miR-495 greatly inhibits cell viability and increases cell apoptosis. Furthermore, our studies demonstrate that PBX3 and MEIS1 are two direct target genes of miR-495, and forced expression of either of them can reverse the effects of miR-495 overexpression on inhibiting cell viability and promoting apoptosis of human MLL-rearranged leukemic cells. Thus, our data indicate that miR-495 likely functions as a tumor suppressor in AML with MLL rearrangements by targeting essential leukemia-related genes.


Clinical Cancer Research | 2008

Histone deacetylase inhibitor romidepsin has differential activity in core binding factor acute myeloid leukemia.

Olatoyosi Odenike; Serhan Alkan; Dorie Sher; John E. Godwin; Dezheng Huo; Stephen J. Brandt; Margaret Green; Jingping Xie; Yanming Zhang; David H. Vesole; Patrick J. Stiff; John J. Wright; Richard A. Larson; Wendy Stock

Purpose: Recruitment of histone deacetylases (HDAC) is a mechanism of transcriptional repression implicated in the differentiation block in acute myeloid leukemia (AML). We hypothesized that the HDAC inhibitor romidepsin could cause transcriptional derepression, up-regulation of specific target genes in AML, and differentiation of the leukemic clone. The primary objectives of the study were to evaluate the safety and efficacy of romidepsin in advanced AML. Experimental Design: Twenty patients were stratified into cohort A or B based on the absence or presence of chromosomal abnormalities known to recruit HDACs, including those involving core binding factor (CBF). Romidepsin was administered i.v. at 13 mg/m2/d on days 1, 8, and 15 of a 28-day cycle. Pharmacodynamic endpoints were evaluated at serial time points. Results: Common adverse effects noted were grade 1 to 2 nausea, anorexia, and fatigue. No objective evidence of antileukemic activity was seen in cohort A. In cohort B, although there were no clinical responses by standard criteria, antileukemic activity was observed in 5 of 7 patients. Two patients had clearance of bone marrow blasts and 3 patients had a >50% decrease in bone marrow blasts. Furthermore, in cohort B, at 24 h, there was a significant increase in MDR1 (P = 0.005), p15 (P = 0.01), and p14 (P < 0.0001) expression. In cohort A, although there was a trend toward up-regulation of MDR1, p15, and p14 expression, these changes were not statistically significant. Conclusion: Romidepsin has differential antileukemic and molecular activity in CBF AML. Development of this agent in CBF AML should focus on combinations that target related mechanisms of gene silencing such as DNA methylation.

Collaboration


Dive into the Yanming Zhang's collaboration.

Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Jianjun Chen

University of Cincinnati

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Zejuan Li

University of Chicago

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge