Zhi Jiang Zang
National University of Singapore
Network
Latest external collaboration on country level. Dive into details by clicking on the dots.
Publication
Featured researches published by Zhi Jiang Zang.
Nature Genetics | 2012
Choon Kiat Ong; Chutima Subimerb; Chawalit Pairojkul; Sopit Wongkham; Ioana Cutcutache; Willie Yu; John R. McPherson; George E. Allen; Cedric Chuan Young Ng; Bernice Huimin Wong; Swe Swe Myint; Vikneswari Rajasegaran; Hong Lee Heng; Anna Gan; Zhi Jiang Zang; Yingting Wu; Jeanie Wu; Ming Hui Lee; Dachuan Huang; Pauline Ong; Waraporn Chan-on; Yun Cao; Chao Nan Qian; Kiat Hon Lim; Aikseng Ooi; Karl Dykema; Kyle A. Furge; Veerapol Kukongviriyapan; Banchob Sripa; Chaisiri Wongkham
Opisthorchis viverrini–related cholangiocarcinoma (CCA), a fatal bile duct cancer, is a major public health concern in areas endemic for this parasite. We report here whole-exome sequencing of eight O. viverrini–related tumors and matched normal tissue. We identified and validated 206 somatic mutations in 187 genes using Sanger sequencing and selected 15 genes for mutation prevalence screening in an additional 46 individuals with CCA (cases). In addition to the known cancer-related genes TP53 (mutated in 44.4% of cases), KRAS (16.7%) and SMAD4 (16.7%), we identified somatic mutations in 10 newly implicated genes in 14.8–3.7% of cases. These included inactivating mutations in MLL3 (in 14.8% of cases), ROBO2 (9.3%), RNF43 (9.3%) and PEG3 (5.6%), and activating mutations in the GNAS oncogene (9.3%). These genes have functions that can be broadly grouped into three biological classes: (i) deactivation of histone modifiers, (ii) activation of G protein signaling and (iii) loss of genome stability. This study provides insight into the mutational landscape contributing to O. viverrini–related CCA.
Genome Biology | 2012
Niranjan Nagarajan; Denis Bertrand; Axel M. Hillmer; Zhi Jiang Zang; Fei Yao; Pierre-Étienne Jacques; Audrey S.M. Teo; Ioana Cutcutache; Zhenshui Zhang; Wah Heng Lee; Yee Yen Sia; Song Gao; Pramila Ariyaratne; Andrea Ho; Xing Yi Woo; Lavanya Veeravali; Choon Kiat Ong; Niantao Deng; Kartiki Vasant Desai; Chiea Chuen Khor; Martin L. Hibberd; Atif Shahab; Jaideepraj Rao; Mengchu Wu; Ming Teh; Feng Zhu; Sze Yung Chin; Brendan Pang; Jimmy By So; Guillaume Bourque
BackgroundGastric cancer is the second highest cause of global cancer mortality. To explore the complete repertoire of somatic alterations in gastric cancer, we combined massively parallel short read and DNA paired-end tag sequencing to present the first whole-genome analysis of two gastric adenocarcinomas, one with chromosomal instability and the other with microsatellite instability.ResultsIntegrative analysis and de novo assemblies revealed the architecture of a wild-type KRAS amplification, a common driver event in gastric cancer. We discovered three distinct mutational signatures in gastric cancer - against a genome-wide backdrop of oxidative and microsatellite instability-related mutational signatures, we identified the first exome-specific mutational signature. Further characterization of the impact of these signatures by combining sequencing data from 40 complete gastric cancer exomes and targeted screening of an additional 94 independent gastric tumors uncovered ACVR2A, RPL22 and LMAN1 as recurrently mutated genes in microsatellite instability-positive gastric cancer and PAPPA as a recurrently mutated gene in TP53 wild-type gastric cancer.ConclusionsThese results highlight how whole-genome cancer sequencing can uncover information relevant to tissue-specific carcinogenesis that would otherwise be missed from exome-sequencing data.
Cancer Research | 2011
Zhi Jiang Zang; Choon Kiat Ong; Ioana Cutcutache; Willie Yu; Shen Li Zhang; Dachuan Huang; Lian Dee Ler; Karl Dykema; Anna Gan; Jiong Tao; Siyu Lim; Yujing Liu; Phillip Andrew Futreal; Heike Grabsch; Kyle A. Furge; Liang Kee Goh; Steve Rozen; Bin Tean Teh; Patrick Tan
Genetic alterations in kinases have been linked to multiple human pathologies. To explore the landscape of kinase genetic variation in gastric cancer (GC), we used targeted, paired-end deep sequencing to analyze 532 protein and phosphoinositide kinases in 14 GC cell lines. We identified 10,604 single-nucleotide variants (SNV) in kinase exons including greater than 300 novel nonsynonymous SNVs. Family-wise analysis of the nonsynonymous SNVs revealed a significant enrichment in mitogen-activated protein kinase (MAPK)-related genes (P < 0.01), suggesting a preferential involvement of this kinase family in GC. A potential antioncogenic role for MAP2K4, a gene exhibiting recurrent alterations in 2 lines, was functionally supported by siRNA knockdown and overexpression studies in wild-type and MAP2K4 variant lines. The deep sequencing data also revealed novel, large-scale structural rearrangement events involving kinases including gene fusions involving CDK12 and the ERBB2 receptor tyrosine kinase in MKN7 cells. Integrating SNVs and copy number alterations, we identified Hs746T as a cell line exhibiting both splice-site mutations and genomic amplification of MET, resulting in MET protein overexpression. When applied to primary GCs, we identified somatic mutations in 8 kinases, 4 of which were recurrently altered in both primary tumors and cell lines (MAP3K6, STK31, FER, and CDKL5). These results demonstrate that how targeted deep sequencing approaches can deliver unprecedented multilevel characterization of a medically and pharmacologically relevant gene family. The catalog of kinome genetic variants assembled here may broaden our knowledge on kinases and provide useful information on genetic alterations in GC.
PLOS ONE | 2014
Maggie Brett; John R. McPherson; Zhi Jiang Zang; Angeline Lai; Ee Shien Tan; Ivy Ng; Lai Choo Ong; Breana Cham; Patrick Tan; Steve Rozen; Ene Choo Tan
Developmental delay and/or intellectual disability (DD/ID) affects 1–3% of all children. At least half of these are thought to have a genetic etiology. Recent studies have shown that massively parallel sequencing (MPS) using a targeted gene panel is particularly suited for diagnostic testing for genetically heterogeneous conditions. We report on our experiences with using massively parallel sequencing of a targeted gene panel of 355 genes for investigating the genetic etiology of eight patients with a wide range of phenotypes including DD/ID, congenital anomalies and/or autism spectrum disorder. Targeted sequence enrichment was performed using the Agilent SureSelect Target Enrichment Kit and sequenced on the Illumina HiSeq2000 using paired-end reads. For all eight patients, 81–84% of the targeted regions achieved read depths of at least 20×, with average read depths overlapping targets ranging from 322× to 798×. Causative variants were successfully identified in two of the eight patients: a nonsense mutation in the ATRX gene and a canonical splice site mutation in the L1CAM gene. In a third patient, a canonical splice site variant in the USP9X gene could likely explain all or some of her clinical phenotypes. These results confirm the value of targeted MPS for investigating DD/ID in children for diagnostic purposes. However, targeted gene MPS was less likely to provide a genetic diagnosis for children whose phenotype includes autism.
Journal of Translational Medicine | 2009
Jit Kong Cheong; Lakshman Gunaratnam; Zhi Jiang Zang; Christopher Maolin Yang; Xiaoming Sun; Susan L Nasr; Khe Guan Sim; Bee Keow Peh; Suhaimi Bin Abdul Rashid; Joseph V. Bonventre; Manuel Salto-Tellez; Stephen I-Hong Hsu
BackgroundMembers of the TRIP-Br/SERTAD family of mammalian transcriptional coregulators have recently been implicated in E2F-mediated cell cycle progression and tumorigenesis. We, herein, focus on the detailed functional characterization of the least understood member of the TRIP-Br/SERTAD protein family, TRIP-Br2 (SERTAD2).MethodsOncogenic potential of TRIP-Br2 was demonstrated by (1) inoculation of NIH3T3 fibroblasts, which were engineered to stably overexpress ectopic TRIP-Br2, into athymic nude mice for tumor induction and (2) comprehensive immunohistochemical high-throughput screening of TRIP-Br2 protein expression in multiple human tumor cell lines and human tumor tissue microarrays (TMAs). Clinicopathologic analysis was conducted to assess the potential of TRIP-Br2 as a novel prognostic marker of human cancer. RNA interference of TRIP-Br2 expression in HCT-116 colorectal carcinoma cells was performed to determine the potential of TRIP-Br2 as a novel chemotherapeutic drug target.ResultsOverexpression of TRIP-Br2 is sufficient to transform murine fibroblasts and promotes tumorigenesis in nude mice. The transformed phenotype is characterized by deregulation of the E2F/DP-transcriptional pathway through upregulation of the key E2F-responsive genes CYCLIN E, CYCLIN A2, CDC6 and DHFR. TRIP-Br2 is frequently overexpressed in both cancer cell lines and multiple human tumors. Clinicopathologic correlation indicates that overexpression of TRIP-Br2 in hepatocellular carcinoma is associated with a worse clinical outcome by Kaplan-Meier survival analysis. Small interfering RNA-mediated (siRNA) knockdown of TRIP-Br2 was sufficient to inhibit cell-autonomous growth of HCT-116 cells in vitro.ConclusionThis study identifies TRIP-Br2 as a bona-fide protooncogene and supports the potential for TRIP-Br2 as a novel prognostic marker and a chemotherapeutic drug target in human cancer.
Cellular Signalling | 2009
Zhi Jiang Zang; Lakshman Gunaratnam; Jit Kong Cheong; Li Yun Lai; Li-Li Hsiao; Eileen O'Leary; Xiaoming Sun; Manuel Salto-Tellez; Joseph V. Bonventre; Stephen I-Hong Hsu
TRIP-Br proteins are a novel family of transcriptional coregulators involved in E2F-mediated cell cycle progression. Three of the four mammalian members of TRIP-Br family, including TRIP-Br1, are known oncogenes. We now report the identification of the Balpha regulatory subunit of serine/threonine protein phosphatase 2A (PP2A) as a novel TRIP-Br1 interactor, based on an affinity binding assay coupled with mass spectrometry. A GST-TRIP-Br1 fusion protein associates with catalytically active PP2A-ABalphaC holoenzyme in vitro. Coimmunoprecipitation confirms this association in vivo. Immunofluorescence staining with a monoclonal antibody against TRIP-Br1 reveals that endogenous TRIP-Br1 and PP2A-Balpha colocalize mainly in the cytoplasm. Consistently, immunoprecipitation followed by immunodetection with anti-phosphoserine antibody suggest that TRIP-Br1 exists in a serine-phosphorylated form. Inhibition of PP2A activity by okadaic acid or transcriptional silencing of the PP2A catalytic subunit by small interfering RNA results in downregulation of total TRIP-Br1 protein levels but upregulation of serine-phosphorylated TRIP-Br1. Overexpression of PP2A catalytic subunit increases TRIP-Br1 protein levels and TRIP-Br1 co-activated E2F1/DP1 transcription. Our data support a model in which association between PP2A-ABalphaC holoenzyme and TRIP-Br1 in vivo in mammalian cells represents a novel mechanism for regulating the level of TRIP-Br1 protooncoprotein.
Journal of Clinical Oncology | 2011
Iain Beehuat Tan; Ioana Cutcutache; Zhi Jiang Zang; Jabed Iqbal; Seow Fong Yap; William Ying Khee Hwang; Wan-Teck Lim; Bin Tean Teh; Steve Rozen; Eng-Huat Tan; Patrick Tan
Case Report A 32-year-old woman was referred for a painless neck lump. A locally advanced tumor was seen on endoscopy, and a biopsy showed a squamous cell carcinoma. Computed tomography imaging revealed a T2N2b irregularly enhancing tumor in the right pyriform sinus with right-sided cervical and retropharyngeal lymphadenopathy (Fig 1, arrow). There were no distant metastases. The patient was enrolled onto a phase II clinical trial evaluating the addition of an epidermal growth factor receptor–targeting monoclonal antibody to concurrent cisplatin/radiation, the standard of care for locally advanced head and neck squamous cell carcinoma (HNSCC). 10 days after receiving a single dose of cisplatin (100 mg/m2) concurrent with daily radiation, she was admitted to the intensive care unit with neutropenic (total white cell count: 0.07 10/L) sepsis with multiorgan failure requiring inotropic and ventilator support. Although initial hemodynamic improvement allowed transfer out from the intensive care unit, she continued to have profound (hemoglobin 7 g/dL, total white count 0.1 10/L, and platelets 100 10/L) and persistent bone marrow failure ( 6 weeks) despite copious blood product support and daily administration of subcutaneous recombinant granulocyte-colony-stimulating factor. During her admission, she developed multiple infective complications including necrotizing enterocolitis, pulmonary abscess, and disseminated fungal infection with aspergilloma in the pons (Fig 2, arrow). A bone marrow biopsy showed a severely hypoplastic marrow with no hematopoiesis, granuloma, nor malignant cells (Fig 3). During her admission, additional family history was provided. The patient’s parents were consanguineous (first cousins). Both her siblings were healthy without major diseases, although her brother was born with an extra thumb. Given the development of HNSCC at a young age in the absence of risk factors, the development of profound and prolonged bone marrow failure induced by a single dose of cisplatin, a family history of consanguineous parents as well as mild morphological features in both the patient (short stature, 146 cm) and her brother (extra thumb), a clinical diagnosis of Fanconi’s anemia (FA) was made. The gold standard diagnostic test for FA is the chromosome breakage test, in which exposure to diepoxybutane (DEB) results in unrepaired DNA double-strand breaks microscopically visible as chromatid breaks and radial chromosomes in nearly all cultured cells. Unfortunately, despite phytohaemagglutinin-stimulated bone marrow culture, insufficient metaphases were available from the pancytopenic patient for full diagnostic work-up, although the few metaphases present were suggestive of increased chromosomal breakage. In the patient’s brother, chromosomal breakage studies using DEB showed a significant level of breakage, diagnostic of FA (Fig 4, Fig 1. Fig 2. JOURNAL OF CLINICAL ONCOLOGY D I A G N O S I S I N O N C O L O G Y VOLUME 29 NUMBER 20 JULY 1
Blood Cancer Journal | 2015
Maria Teresa Voso; Emiliano Fabiani; Zhi Jiang Zang; Luana Fianchi; Giulia Falconi; A Padella; M. Martini; S Li Zhang; Rosaria Santangelo; L. M. Larocca; Marianna Criscuolo; A La Brocca; Ioana Cutcutache; Steve Rozen; G Simonetti; M Manfrini; Giovanni Martinelli; Stefan Hohaus; Giuseppe Leone; Patrick Tan; Daniel G. Tenen
Therapy-related myeloid neoplasms (t-MN) include myelodysplastic syndromes (MDS) and acute myeloid leukemias (AML) occurring as a late effect of chemotherapy and/or radiotherapy for a primary malignancy or for autoimmune diseases.1, 2 The incidence of this complication has been raising in the past years because of the prolonged survival and the higher number of treated patients. Still, <5% of patients exposed to cytotoxic drugs and radiotherapy develop a t-MN, suggesting an underlying individual susceptibility. Primary malignancies most frequently associated to t-MN are breast cancer and lymphoproliferative diseases. Other recurrent clinical characteristics are presence of multiple primary neoplasms in the same individual and cancer familiarity.2 So far, higher frequency of single-nucleotide variants of detoxification and DNA-repair enzymes, alone or in association, have been reported in t-MN, but none has been validated as significant risk factor in large patient groups.3, 4, 5, 6
Cancer Biology & Therapy | 2007
Zhi Jiang Zang; Khe Guan Sim; Jit Kong Cheong; Christopher Maolin Yang; Chui Sun Yap; Stephen I-Hong Hsu
TRIP-Br1 and TRIP-Br2 are potent cell growth promoting factors that function as components of the E2F1/DP1 transcription complex to integrate positive growth signals provided by PHD zinc finger- and/or bromodomain-containing transcription factors. TRIP-Br1 has been demonstrated to be an oncogene. We recently reported that antagonism of the TRIP-Br integrator function by synthetic decoy peptides that compete with TRIP-Br for binding to PHD zinc finger- and/or bromodomain-containing proteins elicit an anti-proliferative effect and induces caspase-3-independent sub-diploidization in cancer cells in vitro. We now demonstrate the chemotherapeutic potential of TRIP-Br decoy peptides for the treatment of cutaneous and intracavitary lesions in vitro as well as in vivo in representative human nasopharyngeal cancer (CNE2), cervical cancer (Ca Ski) and melanoma (MeWo) cancer cell lines. In vitro, BrdU incorporation, colony formation assays and cell cycle analysis confirmed that TRIP-Br decoy peptides possess strong anti-proliferative effects and induce nuclear sub-diploidization in cancer cells. In vivo, NPC2, Ca Ski and MeWo-derived chick embryo chorioallantoic membrane (CAM) tumor xenografts were used to evaluate the effect of topically applied TRIP-Br peptides. Confocal microscopy and flow cytometric analysis demonstrated that cells comprising the tumor xenografts efficiently internalized topically applied FITC-labeled peptides. Fifty µM of TRIP-Br1 decoy peptide significantly suppressed the growth of NPC2-derived human nasopharyngeal tumors, while 50 µM of TRIP-Br2 decoy peptide significantly inhibited tumor growth in all three CAM tumor xenograft models. Two hundred µM of TRIP-Br1 decoy peptide significantly inhibited MeWo-derived tumors. These results suggest that the TRIP-Br integrator function may represent a novel chemotherapeutic target for the treatment of human cutaneous and intracavitary proliferative lesions.
Archive | 2014
Maria Teresa Voso; Emiliano Fabiani; Zhi Jiang Zang; Luana Fianchi; Giulia Falconi; M. Martini; S. L. Zhang; Rosaria Santangelo; L. M. Larocca; Marianna Criscuolo; A. La Brocca; Ioana Cutcutache; Steve Rozen; Stefan Hohaus; Giuseppe Leone; Patrick Tan; Daniel G. Tenen