Network


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

Hotspot


Dive into the research topics where Sai-Juan Chen is active.

Publication


Featured researches published by Sai-Juan Chen.


Nature Genetics | 2011

Exome sequencing identifies somatic mutations of DNA methyltransferase gene DNMT3A in acute monocytic leukemia

Xiao-Jing Yan; Jie Xu; Zhao-Hui Gu; Chun-Ming Pan; Gang Lu; Yang Shen; Jing-Yi Shi; Yong-Mei Zhu; Lin Tang; Xiao-Wei Zhang; Wen-Xue Liang; Jian-Qing Mi; Huai-Dong Song; Ke-Qin Li; Zhu Chen; Sai-Juan Chen

Abnormal epigenetic regulation has been implicated in oncogenesis. We report here the identification of somatic mutations by exome sequencing in acute monocytic leukemia, the M5 subtype of acute myeloid leukemia (AML-M5). We discovered mutations in DNMT3A (encoding DNA methyltransferase 3A) in 23 of 112 (20.5%) cases. The DNMT3A mutants showed reduced enzymatic activity or aberrant affinity to histone H3 in vitro. Notably, there were alterations of DNA methylation patterns and/or gene expression profiles (such as HOXB genes) in samples with DNMT3A mutations as compared with those without such changes. Leukemias with DNMT3A mutations constituted a group of poor prognosis with elderly disease onset and of promonocytic as well as monocytic predominance among AML-M5 individuals. Screening other leukemia subtypes showed Arg882 alterations in 13.6% of acute myelomonocytic leukemia (AML-M4) cases. Our work suggests a contribution of aberrant DNA methyltransferase activity to the pathogenesis of acute monocytic leukemia and provides a useful new biomarker for relevant cases.


The EMBO Journal | 1993

Fusion between a novel Krüppel-like zinc finger gene and the retinoic acid receptor-alpha locus due to a variant t(11;17) translocation associated with acute promyelocytic leukaemia.

Zhu Chen; Nigel J. Brand; Alex Chen; Sai-Juan Chen; Jian-Hua Tong; Samuel Waxman; Arthur Zelent

We have identified a unique case of acute promyelocytic leukaemia (APL) with a t(11;17) reciprocal chromosomal translocation involving the retinoic acid receptor alpha (RAR alpha) and a previously uncharacterized zinc finger gene. As a result of this translocation, mRNAs containing the coding sequences of the new gene, fused in‐frame either upstream of the RAR alpha B region or downstream from the unique A1 and A2 regions of the two major RAR alpha isoforms, are expressed from the rearranged alleles. The above gene, which we have termed PLZF (for promyelocytic leukaemia zinc finger), encodes a potential transcription factor containing nine zinc finger motifs related to the Drosophila gap gene Krüppel and is expressed as at least two isoforms which differ in the sequences encoding the N‐terminal region of the protein. Within the haematopoietic system the PLZF mRNAs were detected in the bone marrow, early myeloid cell lines and peripheral blood mononuclear cells, but not in lymphoid cell lines or tissues. In addition, the PLZF mRNA levels were down‐regulated in NB‐4 and HL‐60 promyelocytic cell lines in response to retinoic acid‐induced granulocytic differentiation and were very low in mature granulocytes. Our results demonstrate for the first time the association of a variant chromosomal translocation involving the RAR alpha gene with APL, further implicating the RAR alpha in leukaemogenesis and also suggesting an important role for PLZF as well as retinoic acid and its receptors in myeloid maturation.


Nature | 2009

The Schistosoma japonicum genome reveals features of host-parasite interplay.

Yan Zhou; Huajun Zheng; Yangyi Chen; Lei Zhang; Kai Wang; Jing Guo; Zhen Huang; Bo Zhang; Wei Huang; Ke Jin; Tonghai Dou; Masami Hasegawa; Wang L; Yuan Zhang; Jie Zhou; Lin Tao; Zhiwei Cao; Yixue Li; Tomas Vinar; Brona Brejova; Daniel G. Brown; Ming Li; David J. Miller; David Blair; Yang Zhong; Zhu Chen; Feng Liu; Wei Hu; Zhi-Qin Wang; Qin-Hua Zhang

Schistosoma japonicum is a parasitic flatworm that causes human schistosomiasis, which is a significant cause of morbidity in China and the Philippines. Here we present a draft genomic sequence for the worm. The genome provides a global insight into the molecular architecture and host interaction of this complex metazoan pathogen, revealing that it can exploit host nutrients, neuroendocrine hormones and signalling pathways for growth, development and maturation. Having a complex nervous system and a well-developed sensory system, S. japonicum can accept stimulation of the corresponding ligands as a physiological response to different environments, such as fresh water or the tissues of its intermediate and mammalian hosts. Numerous proteases, including cercarial elastase, are implicated in mammalian skin penetration and haemoglobin degradation. The genomic information will serve as a valuable platform to facilitate development of new interventions for schistosomiasis control.


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

Dissection of mechanisms of Chinese medicinal formula Realgar-Indigo naturalis as an effective treatment for promyelocytic leukemia.

Lan Wang; Guang-Biao Zhou; Ping Liu; Jun Hong Song; Yang Liang; Xiao Jing Yan; Fang Xu; Bing Shun Wang; Jian-Hua Mao; Zhi Xiang Shen; Sai-Juan Chen; Zhu Chen

To enhance therapeutic efficacy and reduce adverse effects, practitioners of traditional Chinese medicine (TCM) prescribe a combination of plant species/minerals, called formulae, based on clinical experience. Nearly 100,000 formulae have been recorded, but the working mechanisms of most remain unknown. In trying to address the possible beneficial effects of formulae with current biomedical approaches, we use Realgar-Indigo naturalis formula (RIF), which has been proven to be very effective in treating human acute promyelocytic leukemia (APL) as a model. The main components of RIF are realgar, Indigo naturalis, and Salvia miltiorrhiza, with tetraarsenic tetrasulfide (A), indirubin (I), and tanshinone IIA (T) as major active ingredients, respectively. Here, we report that the ATI combination yields synergy in the treatment of a murine APL model in vivo and in the induction of APL cell differentiation in vitro. ATI causes intensified ubiquitination/degradation of promyelocytic leukemia (PML)-retinoic acid receptor α (RARα) oncoprotein, stronger reprogramming of myeloid differentiation regulators, and enhanced G1/G0 arrest in APL cells through hitting multiple targets compared with the effects of mono- or biagents. Furthermore, ATI intensifies the expression of Aquaglyceroporin 9 and facilitates the transportation of A into APL cells, which in turn enhances A-mediated PML-RARα degradation and therapeutic efficacy. Our data also indicate A as the principal component of the formula, whereas T and I serve as adjuvant ingredients. We therefore suggest that dissecting the mode of action of clinically effective formulae at the molecular, cellular, and organism levels may be a good strategy in exploring the value of traditional medicine.


Science | 2010

Arsenic Trioxide Controls the Fate of the PML-RARα Oncoprotein by Directly Binding PML

Xiao-Wei Zhang; Xiao-Jing Yan; Zi-Ren Zhou; Feifei Yang; Ziyu Wu; Hongbin Sun; Wen-Xue Liang; Ai-Xin Song; Valérie Lallemand-Breitenbach; Marion Jeanne; Qun-Ye Zhang; Huai-Yu Yang; Qiu-Hua Huang; Guang-Biao Zhou; Jian-Hua Tong; Yan Zhang; Jihui Wu; Hong-Yu Hu; Sai-Juan Chen; Zhu Chen

Arsenic on the Fingers Arsenic, an ancient drug used in traditional Chinese medicine, has attracted wide interest because it has therapeutic activity in patients with acute promyelocytic leukemia (APL). The drug acts by promoting degradation of an oncogenic protein, PML-RARα, a fusion protein containing sequences from the PML zinc finger protein and retinoic acid receptor α, which is found specifically in APL cells and helps drive their growth. Zhang et al. (p. 240; see the Perspective by Kogan) now explain how arsenic initiates the molecular events leading to PML-RARα degradation. Arsenic was found to bind directly to cysteine residues within zinc finger domains of PML. Arsenic binding then induced oligomerization of PML, which in turn enhanced its association with an enzyme that helps catalyze SUMOylation, a posttranslational modification that can target proteins for degradation. Arsenic, a drug used clinically for leukemia, binds directly to an oncogenic protein, thereby promoting its degradation. Arsenic, an ancient drug used in traditional Chinese medicine, has attracted worldwide interest because it shows substantial anticancer activity in patients with acute promyelocytic leukemia (APL). Arsenic trioxide (As2O3) exerts its therapeutic effect by promoting degradation of an oncogenic protein that drives the growth of APL cells, PML-RARα (a fusion protein containing sequences from the PML zinc finger protein and retinoic acid receptor alpha). PML and PML-RARα degradation is triggered by their SUMOylation, but the mechanism by which As2O3 induces this posttranslational modification is unclear. Here we show that arsenic binds directly to cysteine residues in zinc fingers located within the RBCC domain of PML-RARα and PML. Arsenic binding induces PML oligomerization, which increases its interaction with the small ubiquitin-like protein modifier (SUMO)–conjugating enzyme UBC9, resulting in enhanced SUMOylation and degradation. The identification of PML as a direct target of As2O3 provides new insights into the drug’s mechanism of action and its specificity for APL.


Proceedings of the National Academy of Sciences of the United States of America | 2009

Long-term efficacy and safety of all-trans retinoic acid/arsenic trioxide-based therapy in newly diagnosed acute promyelocytic leukemia

Jiong Hu; Yuan Fang Liu; Chuan Feng Wu; Fang Xu; Zhi Xiang Shen; Yong Mei Zhu; Jun Min Li; Wei Tang; Zhao Wl; Wen Wu; Hui Ping Sun; Qiu Sheng Chen; Bing Chen; Guang-Biao Zhou; Arthur Zelent; Samuel Waxman; Sai-Juan Chen; Zhu Chen

All-trans retinoic acid (ATRA)/arsenic trioxide (ATO) combination-based therapy has benefitted newly diagnosed acute promyelocytic leukemia (APL) in short-term studies, but the long-term efficacy and safety remained unclear. From April 2001, we have followed 85 patients administrated ATRA/ATO with a median follow-up of 70 months. Eighty patients (94.1%) entered complete remission (CR). Kaplan–Meier estimates of the 5-year event-free survival (EFS) and overall survival (OS) for all patients were 89.2% ± 3.4% and 91.7% ± 3.0%, respectively, and the 5-year relapse-free survival (RFS) and OS for patients who achieved CR (n = 80) were 94.8% ± 2.5% and 97.4% ± 1.8%, respectively. Upon ATRA/ATO, prognosis was not influenced by initial white blood cell count, distinct PML-RARα types, or FLT3 mutations. The toxicity profile was mild and reversible. No secondary carcinoma was observed, and 24 months after the last dose of ATRA/ATO, patients had urine arsenic concentrations well below the safety limit. These results demonstrate the high efficacy and minimal toxicity of ATRA/ATO treatment for newly diagnosed APL in long-term follow-up, suggesting a potential frontline therapy for de novo APL.


Leukemia | 2000

Arsenic trioxide-induced apoptosis and differentiation are associated respectively with mitochondrial transmembrane potential collapse and retinoic acid signaling pathways in acute promyelocytic leukemia

Cai X; Shen Yl; Zhu Q; Jia Pm; Yehua Yu; Zhou L; Huang Y; Jiwang Zhang; Shu-Min Xiong; Sai-Juan Chen; Wang Zy; Zhu Chen; Guo-Qiang Chen

Recent studies showed that arsenic trioxide (As2O3) could induce apoptosis and partial differentiation of leukemic promyelocytes. Here, we addressed the possible mechanisms underlying these two different effects. 1.0 μM As2O3-induced apoptosis was associated with condensation of the mitochondrial matrix, disruption of mitochondrial transmembrane potentials (ΔΨm) and activation of caspase-3 in acute promyelocytic leukemia (APL) cells regardless of their sensitivity to all-trans retinoic acid (ATRA). All these effects were inhibited by dithiothreitol (DTT) and enhanced by buthionine sulfoximine (BSO). Furthermore, BSO could also render HL60 and U937 cells, which had the higher cellular catalase activity, sensitive to As2O3-induced apoptosis. Surprisingly, 1.0 μM As2O3 did not induce the ΔΨm collapse and apoptosis, while 0.1 μM As2O3 induced partial differentiation of fresh BM cells from a de novo APL patient. In this study, we also showed that 0.2 mM DTT did not block low-dose As2O3-induced NB4 cell differentiation, and 0.10.5 μM As2O3 did not induce differentiation of ATRA-resistant NB4-derived sublines, which were confirmed by cytomorphology, expression of CD11b, CD33 and CD14 as well as NBT reduction. Another interesting finding was that 0.10.5 μM As2O3 could also induce differentiation-related changes in ATRA-sensitive HL60 cells. However, the differentiation-inducing effect could not be seen in ATRA-resistant HL60 sublines with RARα mutation. Moreover, low-dose As2O3 and ATRA yielded similar gene expression profiles in APL cells. These results encouraged us to hypothesize that As2O3 induces APL cell differentiation through direct or indirect activation of retinoic acid receptor-related signaling pathway(s), while ΔΨm collapse is the common mechanism of As2O3-induced apoptosis.


Blood | 2011

Gene mutation patterns and their prognostic impact in a cohort of 1185 patients with acute myeloid leukemia

Yang Shen; Yong-Mei Zhu; Xing Fan; Jing-Yi Shi; Qin-Rong Wang; Xiao-Jing Yan; Zhao-Hui Gu; Yan-Yan Wang; Bing Chen; Chun-Lei Jiang; Han Yan; Feifei Chen; Chen Hm; Zhu Chen; Jie Jin; Sai-Juan Chen

To evaluate the prognostic value of genetic mutations for acute myeloid leukemia (AML) patients, we examined the gene status for both fusion products such as AML1 (CBFα)-ETO, CBFβ-MYH11, PML-RARα, and MLL rearrangement as a result of chromosomal translocations and mutations in genes including FLT3, C-KIT, N-RAS, NPM1, CEBPA, WT1, ASXL1, DNMT3A, MLL, IDH1, IDH2, and TET2 in 1185 AML patients. Clinical analysis was mainly carried out among 605 cases without recognizable karyotype abnormalities except for 11q23. Of these 605 patients, 452 (74.7%) were found to have at least 1 mutation, and the relationship of gene mutations with clinical outcome was investigated. We revealed a correlation pattern among NPM1, DNMT3A, FLT3, IDH1, IDH2, CEBPA, and TET2 mutations. Multivariate analysis identified DNMT3A and MLL mutations as independent factors predicting inferior overall survival (OS) and event-free survival (EFS), whereas biallelic CEBPA mutations or NPM1 mutations without DNMT3A mutations conferred a better OS and EFS in both the whole group and among younger patients < 60 years of age. The use of molecular markers allowed us to subdivide the series of 605 patients into distinct prognostic groups with potential clinical relevance.


Oncogene | 2001

Arsenic trioxide, a therapeutic agent for APL

Ting-Dong Zhang; Guo-Qiang Chen; Wang Z; Zhen-Yi Wang; Sai-Juan Chen; Zhu Chen

Acute promyelocytic leukemia (APL) is an interesting model in cancer research, because it can respond to the differentiation/apoptosis induction therapy using all-trans retinoic acid (ATRA) and arsenic trioxide (As2O3). Over the past 5 years, it has been well demonstrated that As2O3 induces a high complete remission (CR) rate in both primary and relapsed APL patients (around 85∼90%). The side effects are mild to moderate in relapsed patients, while severe hepatic lesions have been found in some primary cases. After CR obtained in relapsed patients, chemotherapy in combination with As2O3 as post-remission therapy has given better survival than those treated with As2O3 alone. The effect of As2O3 has been shown to be related to the expression of APL-specific PML–RARα oncoprotein, and there is a synergistic effect between As2O3 and ATRA in an APL mouse model. Cell biology studies have revealed that As2O3 exerts dose-dependent dual effects on APL cells. Apoptosis is evident when cells are treated with 0.5∼2.0 μM of As2O3 while partial differentiation is observed using low concentrations (0.1∼0.5 μM) of the drug. The apoptosis-inducing effect is associated with the collapse of mitochondrial transmembrane potentials in a thiol-dependent manner, whereas the mechanisms underlying APL cell differentiation induced by low dose arsenic remain to be explored. Interestingly, As2O3 over a wide range of concentration (0.1∼2.0 μM) induces degradation of a key leukemogenic protein, PML–RARα, as well as the wild-type PML, thus setting up a good example of targeting therapy for human cancers.


Leukemia | 2001

Studies on the clinical efficacy and pharmacokinetics of low-dose arsenic trioxide in the treatment of relapsed acute promyelocytic leukemia: a comparison with conventional dosage

Shen Y; Zhi-Xiang Shen; Hai Yan; Chen J; Zeng Xy; Junmin Li; Li Xs; Wu W; Shu-Min Xiong; Zhao Wl; Tang W; Wu F; Liu Yf; Niu C; Sai-Juan Chen; Zhu Chen

Twenty cases of patients with relapsed acute promyelocytic leukemia (APL) were entered into this study for evaluating the clinical efficacy and pharmacokinetics of low-dose arsenic trioxide (As2O3). As2O3 was given at a daily dose of 0.08 mg/kg intravenously for 28 days. Pharmacokinetic study was carried out in eight patients. 16/20 (80%) patients achieved CR. The occurrence of some toxic events including gastrointestinal disturbance, facial edema and cardiac toxicity seemed reduced in the low-dose group than those in the standard-dose group. Differentiation changes were observed in peripheral blood, as well as in bone marrow (BM). Pharmacokinetic study showed that the plasma concentration increased soon after administration of As2O3 with the peak values of 1.535–3.424 μmol/l. After infusion, the plasma concentration was around 0.1–0.5 μmol/l. The arsenic concentration of the plasma of BM aspirates 24 h after administration in five patients was close to the level needed for differentiation-inducing effect. The estimated 2-year OS and RFS were 61.55 ± 15.79% and 49.11 ± 15.09% respectively, with no difference as compared with those in patients treated with conventional dose (P = 0.2865 and 0.7146, respectively). In conclusion, we demonstrated that low-dose As2O3 had the same effect as the conventional dosage and the mechanism of low-dose arsenic seemed to primarily induce differentiation of APL cells.

Collaboration


Dive into the Sai-Juan Chen's collaboration.

Top Co-Authors

Avatar

Zhu Chen

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Zhi-Xiang Shen

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Bing Chen

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Junmin Li

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Qiu-Hua Huang

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Zhen-Yi Wang

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Guang-Biao Zhou

Chinese Academy of Sciences

View shared research outputs
Top Co-Authors

Avatar

Jian-Qing Mi

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Jing-Yi Shi

Shanghai Jiao Tong University

View shared research outputs
Top Co-Authors

Avatar

Wei-Li Zhao

Shanghai Jiao Tong University

View shared research outputs
Researchain Logo
Decentralizing Knowledge