Network


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

Hotspot


Dive into the research topics where Nellie Yuk Fei Chung is active.

Publication


Featured researches published by Nellie Yuk Fei Chung.


Oncogene | 2010

Minichromosome maintenance proteins 2, 3 and 7 in medulloblastoma: overexpression and involvement in regulation of cell migration and invasion

Kin Mang Lau; Queeny Kwan Yi Chan; Jesse Chung Sean Pang; Kay Ka Wai Li; Walter Wai Yeung; Nellie Yuk Fei Chung; P. C. Lui; Y. S. Tam; Hiu Ming Li; Liangfu Zhou; Yuexiang Wang; Ying Mao; H. K. Ng

Minichromosome maintenance (MCM) proteins 2–7 are important in DNA replication licensing. Functional roles beyond licensing are speculated. In addition, significances in medulloblastoma (MB) remain unclear. In this study, we showed the frequent deregulation of MCM2 and MCM3 expression in 7 MB cell lines and 31 clinical samples. Moreover, DAOY and ONS76 and the clinical samples expressed elevated MCM7 transcripts with genomic gain of the gene. Immunopositivity restricted to tumor cells was found in 41, 37 and 53 out of 73 MB cases for MCM2, MCM3 and MCM7, respectively. High-MCM3 expression was associated with poor prognosis. Knockdowns of these MCMs significantly inhibited anchorage-dependent and -independent MB cell growth. The inhibition of MCM3 expression by small interfering RNA knockdown was related to G1 arrest with reduced cyclin A expression, whereas the MCM2- and MCM7-knocked-down cells arrested at G2/M with increased cyclin A expression. Interestingly, we demonstrated the links of these MCMs with cell migration and invasion using wound-healing and Transwell migration/invasion assays. Exogenous overexpression of MCM2, MCM3 and MCM7 increased anchorage-independent cell growth, and also cell migration and invasion capabilities in MB cells. The knockdown reduced the number of filopodial cells and the cells with intense stress fibers by blocking cdc42 and Rho activation. Taken together, deregulation of MCM2, MCM3 and MCM7 expression might be involved in MB tumorigenesis and we revealed undefined roles of these MCMs in control of MB cell migration and invasion.


Modern Pathology | 2015

TERT promoter mutations contribute to subset prognostication of lower-grade gliomas

Aden Ka-Yin Chan; Yu Yao; Zhenyu Zhang; Nellie Yuk Fei Chung; Joseph Shu Ming Liu; Kay Ka Wai Li; Zhifeng Shi; Danny Tat Ming Chan; Wai Sang Poon; Liangfu Zhou; Ho Keung Ng

Recurrent mutations in the promoter region of telomerase reverse transcriptase (TERT) have been found in various cancers including diffuse gliomas. Mutations lead to TERT upregulation and are associated with aggressive clinical behavior in glioblastomas. However, the clinical significance of TERT promoter mutations in lower-grade gliomas remains undetermined. The aim of this study is to evaluate the status of TERT promoter and the respective prognostic significance in a cohort of 237 lower-grade gliomas comprising grades II and III astrocytomas, oligodendrogliomas, and oligoastrocytomas. Mutually exclusive mutations in TERT promoter, C228T and C250T, were identified in 16/105 (15%) diffuse astrocytomas, 16/63 (25%) anaplastic astrocytomas, 13/18 (72%) oligodendrogliomas, 3/3 (100%) anaplastic oligodendrogliomas, 17/45 (38%) oligoastrocytomas, and 2/3 (67%) anaplastic oligoastrocytomas. Mutations co-occurred with 1p/19q codeletion (P<0.001) and are associated with oligodendroglial histology (P<0.001). Kaplan–Meier’s survival analysis showed that TERT promoter mutation (P=0.037), Isocitrate dehydrogenase (IDH) mutation (P<0.001), and 1p/19q codeletion (P<0.001) were associated with favorable overall survival (OS). In the subset of 116 IDH-mutated lower-grade gliomas lacking 1p/19q codeletion, 19 TERT promoter-mutated tumors exhibited longer progression-free survival (PFS) (P=0.027) and OS (P=0.004). Consistent with this observation, in the subset of 97 IDH-mutated astrocytomas, 14 TERT promoter-mutated tumors showed longer PFS (P=0.001) and OS (P=0.001). In contrast, among the subset of 74 IDH wild-type lower-grade gliomas with intact 1p/19q, TERT promoter mutation was associated with shorter PFS (P=0.001) and OS (P=0.001). Similarly, in the subset of 65 IDH wild-type astrocytomas, 16 TERT promoter-mutated tumors exhibited unfavorable PFS (P=0.007) and OS (P=0.008). Our results indicate that when combined with IDH status, TERT promoter mutation contributes to prognostic subgroups of lower-grade astrocytic tumors or 1p/19q intact lower-grade gliomas and this may further refine future molecular classification of lower-grade gliomas.


Modern Pathology | 2014

Loss of CIC and FUBP1 expressions are potential markers of shorter time to recurrence in oligodendroglial tumors

Aden Ka-Yin Chan; Jesse Pang; Nellie Yuk Fei Chung; Kay Ka Wai Li; Wai Sang Poon; Danny Tat Ming Chan; Zhifeng Shi; Liang Chen; Liangfu Zhou; Ho Keung Ng

Combined deletion of chromosomes 1p and 19q is a prognostic marker in oligodendroglial tumors. Recent studies in oligodendroglial tumors have unveiled recurrent mutations of CIC (homolog of Drosophila capicua) and FUBP1 (far upstream element binding protein 1) that are located on 19q13 and 1p31, respectively. However, the impact of CIC and FUBP1 mutations on their protein expressions has not been examined. The aims of this study were to correlate the expression patterns of CIC and FUBP1 with their mutation profiles and to evaluate the clinical relevance of these molecular markers in 55 oligodendroglial tumors diagnosed in 47 adult patients. Using direct sequencing, somatic mutations of CIC and FUBP1 were identified in 47% (22/47) and 16% (7/45) of oligodendroglial tumors, respectively. Immunohistochemical analysis revealed loss of CIC or FUBP1 protein expression in 36% (20/55) and 16% (9/55) of oligodendroglial tumors examined. Somatic mutation was significantly associated with absent protein expression for both genes (CIC, P=0.01; FUBP1, P=0.00001). Four tumors with undetectable CIC mutations exhibited absent CIC expression, suggesting that CIC inactivation could be mediated by mechanisms other than mutations and genomic loss. Univariate survival analysis revealed that 1p/19q codeletion was significantly associated with overall survival (P=0.05). Loss of CIC expression was significantly correlated with shorter progression-free survival (P=0.03), whereas CIC alteration (mutation or null expression) with worse overall survival (P=0.05). Absent FUBP1 expression was linked with unfavorable progression-free survival (P=0.02) and overall survival (P=0.01). In 16 tumors with 1p/19q codeletion, CIC mutation was associated with unfavorable survival (P=0.01). There was a correlation between lack of CIC or FUBP1 expression and poor progression-free survival (P=0.004; P=0.0003). No molecular markers showed association with survival in oligodendroglial tumors lacking 1p/19q codeletion. We conclude that absent CIC and FUBP1 expressions are potential markers of shorter time to recurrence and CIC mutation a potential marker of worse prognosis, especially in tumors carrying 1p/19q codeletion.


Oncotarget | 2016

Biomarker-based prognostic stratification of young adult glioblastoma

Rui Qi Zhang; Zhifeng Shi; Hong Chen; Nellie Yuk Fei Chung; Zi Yin; Kay Ka Wai Li; Danny Tat Ming Chan; Wai Sang Poon; Jinsong Wu; Liangfu Zhou; Aden Ka-Yin Chan; Ying Mao; Ho Keung Ng

While the predominant elderly and the pediatric glioblastomas have been extensively investigated, young adult glioblastomas were understudied. In this study, we sought to stratify young adult glioblastomas by BRAF, H3F3A and IDH1 mutations and examine the clinical relevance of the biomarkers. In 107 glioblastomas aged from 17 to 35 years, mutually exclusive BRAF-V600E (15%), H3F3A-K27M (15.9%), H3F3A-G34R/V (2.8%) and IDH1-R132H (16.8%) mutations were identified in over half of the cases. EGFR amplification and TERTp mutation were only detected in 3.7% and 8.4% in young adult glioblastomas, respectively. BRAF-V600E identified a clinically favorable subset of glioblastomas with younger age, frequent CDKN2A homozygous deletion, and was more amendable to surgical resection. H3F3A-K27M mutated glioblastomas were tightly associated with midline locations and showed dismal prognosis. IDH1-R132H was associated with older age and favorable outcome. Interestingly, tumors with positive PDGFRA immunohistochemical expression exhibited poorer prognosis and identified an aggressive subset of tumors among K27M mutated glioblastomas. Combining BRAF, H3F3A and IDH1 mutations allowed stratification of young adult glioblastomas into four prognostic subgroups. In summary, our study demonstrates the clinical values of stratifying young adult glioblastomas with BRAF, H3F3A and IDH1 mutations, which has important implications in refining prognostic classification of glioblastomas.


Brain Pathology | 2010

KIAA0495/PDAM Is frequently downregulated in oligodendroglial tumors and its knockdown by siRNA induces cisplatin resistance in glioma cells

Jesse Chung Sean Pang; Kay Ka Wai Li; Kin Mang Lau; Yeung Lam Ng; John Wong; Nellie Yuk Fei Chung; Hiu Ming Li; Yiu-Loon Chui; Vivian Wai Yan Lui; Zhong Ping Chen; Danny Tat Ming Chan; Wai Sang Poon; Yin Wang; Yin Mao; Liangfu Zhou; Ho Keung Ng

Co‐deletion of chromosomes 1p and 19q is a common event in oligodendroglial tumors (OTs), suggesting the presence of OT‐related genes. The aim of this study was to identify the target genes residing in the minimally deleted regions on chromosome 1p36.31–p36.32 that might be involved in OTs. A novel gene KIAA0495/p53‐dependent apoptosis modulator (PDAM) was found frequently deregulated, with 37 of 58 (63.8%) OTs examined showing reduced expression compared with normal brain. Chromosome 1p loss and epigenetic modifications were the major mechanisms contributing to PDAM downregulation. The role of PDAM in chemosensitivity was also evaluated. PDAM knockdown had no effect on sensitivity to vincristine, lomustine, temozolomide and paclitaxel, but could induce cisplatin resistance in glioma cells harboring wild‐type p53. B‐cell CCL/lymphoma 2 (BCL2)‐like 1 (BCL2L1) exhibited significant upregulation, while BCL2 showed partial derepression in PDAM‐silenced cells after cisplatin treatment, suggesting that alteration of anti‐apoptotic genes contributed in part to cisplatin resistance. Knockdown of BCL2L1 abrogated the induced cisplatin‐resistant phenotype. Moreover, our data suggested that PDAM might function as a non‐protein‐coding RNA. Collectively, these findings suggest that PDAM deregulation may play a role in OT development and that PDAM may possess the capacity to modulate apoptosis via regulation of p53‐dependent anti‐apoptotic genes.


International Journal of Cancer | 2007

EMP3 overexpression is associated with oligodendroglial tumors retaining chromosome arms 1p and 19q

Kay Ka Wai Li; Jesse Chung Sean Pang; Nellie Yuk Fei Chung; Yeung Lam Ng; Norman Hok Ling Chan; Liangfu Zhou; Wai Sang Poon; Ho Keung Ng

The epithelial membrane protein 3 (EMP3) gene located on chromosome 19q13 has been implicated as a candidate tumor suppressor gene (TSG) in neuroblastomas and gliomas. The aim of this study was to investigate whether EMP3 is involved in oligodendroglial tumors (OTs), which frequently carry combined chromosomes 1p and 19q deletion. We first investigated the transcript level of EMP3 in a cohort of 57 OTs by quantitative real‐time RT‐PCR. Our results showed that 10 (18%) tumors had reduced EMP3 expression level compared to normal brains. Six of these tumors carried chromosome 19q13 deletion but no statistical correlation was found between the 2 parameters. Intriguingly, a similar proportion (11 of 57, 19%) of tumors displayed EMP3 overexpression, with 8 of them having transcript level >10‐fold higher than normal brain. All 11 OTs retained chromosomes 1p36 and 19q13, and a significant association was found between EMP3 overexpression and balanced chromosomes 1p36 and 19q13 (p = 0.004). The methylation status of EMP3 was evaluated by bisulfite sequencing in 29 OTs with diverse expression levels. All tumors except 3 showed aberrant methylation of EMP3 and no correlation was observed between transcript level and methylation status, suggesting that methylation alone does not mediate transcriptional down‐regulation of EMP3 in OTs. In conclusion, our study demonstrates that EMP3 overexpression is involved in OTs retaining chromosomes 1p and 19q and does not support EMP3 as the target TSG on chromosome 19q13 in OTs.


Acta Neuropathologica | 2006

Rare mutation of PIK3CA in meningiomas

Jesse Chung Sean Pang; Nellie Yuk Fei Chung; Norman Hok Ling Chan; Wai Sang Poon; Teresa M. M. Thomas; Ho Keung Ng

The phosphoinositide 3-kinase (PI3K)/AKT signaling pathway plays a pivotal role in diverse cellular processes, such as proliferation, survival, and cytoskeletal rearrangement [reviewed in 9]. Anomalous activation of this pathway has been implicated in the development and progression of a wide variety of human cancers. For instance, inactivation of PTEN, an antagonist of PI3K, is frequently detected in glioblastomas, whereas activation of AKT2, a downstream effector of PI3K, is reported in breast and ovarian tumors. The PI3K/AKT pathway can also be activated by gene amplification and/or overexpression of PIK3CA, which encodes the p110a catalytic subunit of the class IA PI3K [2, 6]. Recently, Samuels et al. reported that somatic mutation is an alternative mechanism for PIK3CA activation and such genetic aberration has been detected in colorectal, gastric, breast, and lung tumors and glioblastomas [8]. About 80% of the base alterations are found clustered in the helical (exon 9) and kinase (exon 20) domains of PIK3CA. In this study, we investigated whether somatic mutation of PIK3CA was involved in meningiomas. Using direct sequencing, we screened for base alterations at the mutational hotspots of PIK3CA, exons 9 and 20, as well as on exon 1, where somatic mutation has been reported in brain tumor [1]. A series of 78 meningiomas (26 grade I, 33 grade II, and 19 grade III), classified according to the current World Health Organization criteria, were examined. Five of these tumors (one grade I, three grade II, and one grade III) were recurrences. Our mutation analysis identified one base substitution, A3140G, with a predicted amino acid change from histidine to arginine at codon 1047 (H1047R) of exon 20 in a primary grade III meningioma (Fig. 1). Such base change was not observed in patient’s peripheral lymphocytes, indicating that it was a somatic alteration. The patient was a 71-year-old female, presented with expressive dysphasia and cognitive dysfunction. Histologic examination of the tumor revealed frank brain invasion, numerous mitoses, and high MIB1 labeling index. The tumor, located in the left temporal lobe, was debulked. The patient died 10 months after initial diagnosis. In addition, our previous methylation analysis indicated that this malignant tumor also had promoter hypermethylation of RB1 [5]. No base alteration on either exon 1 or exon 9 of PIK3CA was detected in the series examined. The H1047R mutation maps to the kinase domain of PIK3CA and the amino acid residue involved is highly conserved through evolution. Previous studies showed that the H1047R mutant protein had elevated kinase activity in vitro [4, 8]. Forced expression of the mutant gene in chicken embryo fibroblasts led to cellular transformation with high efficiency [4]. Moreover, treatment of breast tumor cell lines carrying various PIK3CA mutations, including H1047R, with PI3K inhibitor resulted in decreased proliferation and increased apoptosis [10]. Together these results strongly suggest that the H1047R mutation of PIK3CA is oncogenic. Mawrin et al. recently demonstrated higher levels of activated, phosphorylated Akt in atypical and malignant meningiomas compared with benign tumors [7]. Their results suggest that activation of the PI3K/AKT signaling contributes to the aggressive behavior of malignant meningiomas. Another component of the PI3K/ AKT pathway reported altered in meningiomas is This study was supported by the Research Grant Council of Hong Kong, Hong Kong, China


Acta Neuropathologica | 2012

Overexpression of HMGA1 deregulates tumor growth via cdc25A and alters migration/invasion through a cdc25A-independent pathway in medulloblastoma

Kin Mang Lau; Queeny Kwan Yi Chan; Jesse Chung Sean Pang; Fanny Man-Ting Ma; Kay K.W. Li; Walter Wai Yeung; Alfred S.L. Cheng; Hai Feng; Nellie Yuk Fei Chung; Hiu Ming Li; Liangfu Zhou; Yin Wang; Ying Mao; Ho Keung Ng

Overexpression of high mobility group AT-hook 1 (HMGA1) is common in human cancers. Little is known about the mechanisms underlying its deregulation and downstream targets, and information about its clinical and biological significance in medulloblastoma (MB) is lacking. Here, we demonstrated frequent genomic gain at 6p21.33–6p21.31 with copy number increase leading to overexpression of HMGA1 in MB. The overexpression correlated with a high proliferation index and poor prognosis. Moreover, we found that hsa-miR-124a targeted 3′UTR of HMGA1 and negatively modulated the expression in MB cells, indicating that loss/downregulation of hsa-miR-124a reported in our previous study could contribute to the overexpression. Regarding the biological significance of HMGA1, siRNA knockdown and ectopic expression studies revealed the crucial roles of HMGA1 in controlling MB cell growth and migration/invasion through modulation of apoptosis and formation of filopodia and stress fibers, respectively. Furthermore, we identified cdc25A as a target of HMGA1 and showed that physical interaction between HMGA1 and the cdc25A promoter is required for transcriptional upregulation. In clinical samples, HMGA1 and cdc25A were concordantly overexpressed. Functionally, cdc25A is involved in the HMGA1-mediated control of MB cell growth. Finally, netropsin, which competes with HMGA1 in DNA binding, reduced the expression of cdc25A by suppression of its promoter activity and inhibited in vitro and in vivo intracranial MB cell growth. In conclusion, our results delineate the mechanisms underlying the deregulation and reveal the functional significance of HMGA1 in controlling MB cell growth and migration/invasion. Importantly, the results highlight the therapeutic potential of targeting HMGA1 in MB patients.


Oncotarget | 2016

Not all 1p/19q non-codeleted oligodendroglial tumors are astrocytic

Yan Xi Li; Zhifeng Shi; Abudumijiti Aibaidula; Hong Chen; Qisheng Tang; Kay Ka Wai Li; Nellie Yuk Fei Chung; Danny Tat Ming Chan; Wai Sang Poon; Ying Mao; Jinsong Wu; Liangfu Zhou; Aden Ka-Yin Chan; Ho Keung Ng

Although 1p/19q codeletion is the genetic hallmark defining oligodendrogliomas, approximately 30-40% of oligodendroglial tumors have intact 1p/19q in the literature and they demonstrate a worse prognosis. This group of 1p/19q intact oligodendroglial tumors is frequently suggested to be astrocytic in nature with TP53 and ATRX mutations but actually remains under-investigated. In the present study, we provided evidence that not all 1p/19q intact oligodendroglial tumors are astrocytic through histologic and molecular approaches. We examined 1p/19q status by FISH in a large cohort of 337 oligodendroglial tumors and identified 39.8% lacking 1p/19q codeletion which was independently associated with poor prognosis. Among this 1p/19q intact oligodendroglial tumor cohort, 58 cases demonstrated classic oligodendroglial histology which showed older patient age, better prognosis, association with grade III histology, PDGFRA expression, TERTp mutation, as well as frequent IDH mutation. More than half of the 1p/19q intact oligodendroglial tumors showed lack of astrocytic defining markers, p53 expression and ATRX loss. TP53 mutational analysis was additionally conducted in 45 cases of the 1p/19q intact oligodendroglial tumors. Wild-type TP53 was detected in 71.1% of cases which was associated with classic oligodendroglial histology. Importantly, IDH and TERTp co-occurred in 75% of 1p/19q intact, TP53 wild-type oligodendrogliomas, highlighting the potential of the co-mutations in assisting diagnosis of oligodendrogliomas in tumors with clear cell morphology and non-codeleted 1p/19q status. In summary, our study demonstrated that not all 1p/19q intact oligodendroglial tumors are astrocytic and co-evaluation of IDH and TERTp mutation could potentially serve as an adjunct for diagnosing 1p/19q intact oligodendrogliomas.


Acta Neuropathologica | 2018

Pediatric low-grade gliomas can be molecularly stratified for risk

Rui Ryan Yang; Abudumijiti Aibaidula; Wei wei Wang; Aden Ka-Yin Chan; Zhi feng Shi; Zhen yu Zhang; Danny Tat Ming Chan; Wai Sang Poon; Xian zhi Liu; Wen cai Li; Rui qi Zhang; Yan Xi Li; Nellie Yuk Fei Chung; Hong Chen; Jingsong Wu; Liangfu Zhou; Kay Ka Wai Li; Ho Keung Ng

Pediatric low-grade gliomas (PLGGs) consist of a number of entities with overlapping histological features. PLGGs have much better prognosis than the adult counterparts, but a significant proportion of PLGGs suffers from tumor progression and recurrence. It has been shown that pediatric and adult low-grade gliomas are molecularly distinct. Yet the clinical significance of some of newer biomarkers discovered by genomic studies has not been fully investigated. In this study, we evaluated in a large cohort of 289 PLGGs a list of biomarkers and examined their clinical relevance. TERT promoter (TERTp), H3F3A and BRAF V600E mutations were detected by direct sequencing. ATRX nuclear loss was examined by immunohistochemistry. CDKN2A deletion, KIAA1549-BRAF fusion, and MYB amplification were determined by fluorescence in situ hybridization (FISH). TERTp, H3F3A, and BRAF V600E mutations were identified in 2.5, 6.4, and 7.4% of PLGGs, respectively. ATRX loss was found in 4.9% of PLGGs. CDKN2A deletion, KIAA1549-BRAF fusion and MYB amplification were detected in 8.8, 32.0 and 10.6% of PLGGs, respectively. Survival analysis revealed that TERTp mutation, H3F3A mutation, and ATRX loss were significantly associated with poor PFS (p < 0.0001, p < 0.0001, and p = 0.0002) and OS (p < 0.0001, p < 0.0001, and p < 0.0001). BRAF V600E was associated with shorter PFS (p = 0.011) and OS (p = 0.032) in a subset of PLGGs. KIAA1549-BRAF fusion was a good prognostic marker for longer PFS (p = 0.0017) and OS (p = 0.0029). MYB amplification was also a favorable marker for a longer PFS (p = 0.040). Importantly, we showed that these molecular biomarkers can be used to stratify PLGGs into low- (KIAA1549-BRAF fusion or MYB amplification), intermediate-I (BRAF V600E and/or CDKN2A deletion), intermediate-II (no biomarker), and high-risk (TERTp or H3F3A mutation or ATRX loss) groups with distinct PFS (p < 0.0001) and OS (p < 0.0001). This scheme should aid in clinical decision-making.

Collaboration


Dive into the Nellie Yuk Fei Chung's collaboration.

Top Co-Authors

Avatar

Ho Keung Ng

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Wai Sang Poon

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Aden Ka-Yin Chan

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Danny Tat Ming Chan

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Kay Ka Wai Li

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar

Jesse Chung Sean Pang

The Chinese University of Hong Kong

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge