Network


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

Hotspot


Dive into the research topics where Hsiang-Fu Kung is active.

Publication


Featured researches published by Hsiang-Fu Kung.


Molecular Biology of the Cell | 2011

MiR-637 maintains the balance between adipocytes and osteoblasts by directly targeting Osterix

Jin Fang Zhang; Wei Ming Fu; Ming Liang He; Hua Wang; Wei Mao Wang; Shi Cang Yu; Xiu Wu Bian; Jin Zhou; Marie C. Lin; Gang Lu; Wai Sang Poon; Hsiang-Fu Kung

Human mesenchymal stem cells are adipoosteogenic progenitors that can differentiate into osteoblasts or adipocytes. A balance between osteogenesis and adipogenesis must be maintained in the growth of connective dynamic tissue. In our study, miR-637 was found to function as the mediator for maintaining the balance of the microenvironment and regulating differentiation.


Cancer Research | 2013

β-Catenin/POU5F1/SOX2 transcription factor complex mediates IGF-I receptor signaling and predicts poor prognosis in lung adenocarcinoma.

Chuan Xu; Dan Xie; Shi Cang Yu; Xiao-jun Yang; Li Ru He; Jing Yang; Yi Fang Ping; Bin Wang; Lang Yang; Sen Lin Xu; Qing Liang Wang; Wen Juan Fu; Qing Liu; Cheng Qian; You hong Cui; Jeremy N. Rich; Hsiang-Fu Kung; Xia Zhang; Xiu Wu Bian

Cancer stem-like cells (CSLC) are crucial in tumor initiation and progression; however, the underlying mechanism for the self-renewal of cancer cells remains undefined. In the study, immunohistochemical analysis of specimens freshly excised from patients with lung adenocarcinoma showed that high expression of insulin-like growth factor I receptor (IGF-IR) in lung adenocarcinoma cells was positively correlated with the expressions of cancer stem cell markers CD133 and aldehyde dehydrogenase 1 family member A1 (ALDH1A1). IGF-IR activation enhanced POU class 5 homeobox 1 (POU5F1) expression on human lung adenocarcinoma stem-like cells (LACSLC) through PI3K/AKT/GSK3β/β-catenin cascade. POU5F1 could form a novel complex with β-catenin and SOX2 to bind Nanog promoter for transcription to maintain self-renewal of LACSLCs, which was dependent on the functional IGF-IR. Genetic and pharmacologic inhibition of IGF-IR abrogated LACSLC capabilities for self-renewal and tumorigenicity in vitro. In an in vivo xenograft tumor model, knockdown of either IGF-IR or POU5F1 impeded tumorigenic potentials of LACSLCs. By analyzing pathologic specimens excised from 200 patients with lung adenocarcinoma, we found that colocalization of highly expressed IGF-IR with β-catenin and POU5F1 predicted poor prognosis. Taken together, we show that IGF-IR-mediated POU5F1 expression to form a complex with β-catenin and SOX2 is crucial for the self-renewal and oncogenic potentials of LACSLCs, and the integrative clinical detection of the expressions of IGF-IR, β-catenin, and POU5F1 is indicatory for predicting prognosis in the patients of lung adenocarcinoma.


The Journal of Pathology | 2017

High-mobility group box 1 released by autophagic cancer-associated fibroblasts maintains the stemness of luminal breast cancer cells: Cross-talk between autophagic CAFs and BCICs

Xi-Long Zhao; Yong Lin; Jun Jiang; Zhuo Tang; Shuai Yang; Lu Lu; Yan Liang; Xue Liu; Jiao Tan; Xu-Gang Hu; Qin Niu; Wen-Juan Fu; Ze‐Xuan Yan; De-Yu Guo; Yi-Fang Ping; Ji Ming Wang; Xia Zhang; Hsiang-Fu Kung; Xiu-wu Bian; Xiao-hong Yao

Cancer stem cells/cancer‐initiating cells (CICs) and their microenvironmental niche play a vital role in malignant tumour recurrence and metastasis. Cancer‐associated fibroblasts (CAFs) are major components of the niche of breast cancer‐initiating cells (BCICs), and their interactions may profoundly affect breast cancer progression. Autophagy has been considered to be a critical process for CIC maintenance, but whether it is involved in the cross‐talk between CAFs and CICs to affect tumourigenesis and pathological significance has not been determined. In this study, we found that the presence of CAFs containing high levels of microtubule‐associated protein 1 light chain 3 (LC3II), a marker of autophagosomes, was associated with more aggressive luminal human breast cancer. CAFs in human luminal breast cancer tissues with high autophagy activity enriched BCICs with increased tumourigenicity. Mechanistically, autophagic CAFs released high‐mobility group box 1 (HMGB1), which activated its receptor, Toll‐like receptor (TLR) 4, expressed by luminal breast cancer cells, to enhance their stemness and tumourigenicity. Furthermore, immunohistochemistry of 180 luminal breast cancers revealed that high LC3II/TLR4 levels predicted an increased relapse rate and a poorer prognosis. Our findings demonstrate that autophagic CAFs play a critical role in promoting the progression of luminal breast cancer through an HMGB1–TLR4 axis, and that both autophagy in CAFs and TLR4 on breast cancer cells constitute potential therapeutic targets. Copyright


The Journal of Pathology | 2017

High‐mobility group box 1 released by autophagic cancer‐associated fibroblasts maintains the stemness of luminal breast cancer cells

Xi-Long Zhao; Yong Lin; Jun Jiang; Zhuo Tang; Shuai Yang; Lu Lu; Yan Liang; Xue Liu; Jiao Tan; Xu-Gang Hu; Qin Niu; Wen-Juan Fu; Ze‐Xuan Yan; De-Yu Guo; Yi-Fang Ping; Ji Ming Wang; Xia Zhang; Hsiang-Fu Kung; Xiu-wu Bian; Xiao-hong Yao

Cancer stem cells/cancer‐initiating cells (CICs) and their microenvironmental niche play a vital role in malignant tumour recurrence and metastasis. Cancer‐associated fibroblasts (CAFs) are major components of the niche of breast cancer‐initiating cells (BCICs), and their interactions may profoundly affect breast cancer progression. Autophagy has been considered to be a critical process for CIC maintenance, but whether it is involved in the cross‐talk between CAFs and CICs to affect tumourigenesis and pathological significance has not been determined. In this study, we found that the presence of CAFs containing high levels of microtubule‐associated protein 1 light chain 3 (LC3II), a marker of autophagosomes, was associated with more aggressive luminal human breast cancer. CAFs in human luminal breast cancer tissues with high autophagy activity enriched BCICs with increased tumourigenicity. Mechanistically, autophagic CAFs released high‐mobility group box 1 (HMGB1), which activated its receptor, Toll‐like receptor (TLR) 4, expressed by luminal breast cancer cells, to enhance their stemness and tumourigenicity. Furthermore, immunohistochemistry of 180 luminal breast cancers revealed that high LC3II/TLR4 levels predicted an increased relapse rate and a poorer prognosis. Our findings demonstrate that autophagic CAFs play a critical role in promoting the progression of luminal breast cancer through an HMGB1–TLR4 axis, and that both autophagy in CAFs and TLR4 on breast cancer cells constitute potential therapeutic targets. Copyright


The Journal of Pathology | 2017

HMGB1 released by autophagic cancer‐associated fibroblasts maintains the stemness of luminal breast cancer cells

Xi-Long Zhao; Yong Lin; Jun Jiang; Zhuo Tang; Shuai Yang; Lu Lu; Yan Liang; Xue Liu; Jiao Tan; Xu-Gang Hu; Qin Niu; Wen-Juan Fu; Ze‐Xuan Yan; De-Yu Guo; Yi-Fang Ping; Ji Ming Wang; Xia Zhang; Hsiang-Fu Kung; Xiu-wu Bian; Xiao-hong Yao

Cancer stem cells/cancer‐initiating cells (CICs) and their microenvironmental niche play a vital role in malignant tumour recurrence and metastasis. Cancer‐associated fibroblasts (CAFs) are major components of the niche of breast cancer‐initiating cells (BCICs), and their interactions may profoundly affect breast cancer progression. Autophagy has been considered to be a critical process for CIC maintenance, but whether it is involved in the cross‐talk between CAFs and CICs to affect tumourigenesis and pathological significance has not been determined. In this study, we found that the presence of CAFs containing high levels of microtubule‐associated protein 1 light chain 3 (LC3II), a marker of autophagosomes, was associated with more aggressive luminal human breast cancer. CAFs in human luminal breast cancer tissues with high autophagy activity enriched BCICs with increased tumourigenicity. Mechanistically, autophagic CAFs released high‐mobility group box 1 (HMGB1), which activated its receptor, Toll‐like receptor (TLR) 4, expressed by luminal breast cancer cells, to enhance their stemness and tumourigenicity. Furthermore, immunohistochemistry of 180 luminal breast cancers revealed that high LC3II/TLR4 levels predicted an increased relapse rate and a poorer prognosis. Our findings demonstrate that autophagic CAFs play a critical role in promoting the progression of luminal breast cancer through an HMGB1–TLR4 axis, and that both autophagy in CAFs and TLR4 on breast cancer cells constitute potential therapeutic targets. Copyright


Journal of Genetics and Genomics | 2017

A glycolysis-based ten-gene signature correlates with the clinical outcome, molecular subtype and IDH1 mutation in glioblastoma

Cong Chen; Yu Shi; Yong Li; Zhi-Cheng He; Kai Zhou; Xiao-Ning Zhang; Kai-Di Yang; Jin-rong Wu; Hsiang-Fu Kung; Yi-Fang Ping; Xiu-wu Bian

Reprogrammed metabolism is a hallmark of cancer. Glioblastoma (GBM) tumor cells predominantly utilize aerobic glycolysis for the biogenesis of energy and intermediate nutrients. However, in GBM, the clinical significance of glycolysis and its underlying relations with the molecular features such as IDH1 mutation and subtype have not been elucidated yet. Herein, based on glioma datasets including TCGA (The Cancer Genome Atlas), REMBRANDT (Repository for Molecular Brain Neoplasia Data) and GSE16011, we established a glycolytic gene expression signature score (GGESS) by incorporating ten glycolytic genes. Then we performed survival analyses and investigated the correlations between GGESS and IDH1 mutation as well as the molecular subtypes in GBMs. The results showed that GGESS independently predicted unfavorable prognosis and poor response to chemotherapy of GBM patients. Notably, GGESS was high in GBMs of mesenchymal subtype but low in IDH1-mutant GBMs. Furthermore, we found that the promoter regions of tumor-promoting glycolytic genes were hypermethylated in IDH1-mutant GBMs. Finally, we found that high GGESS also predicted poor prognosis and poor response to chemotherapy when investigating IDH1-wildtype GBM patients only. Collectively, glycolysis represented by GGESS predicts unfavorable clinical outcome of GBM patients and is closely associated with mesenchymal subtype and IDH1 mutation in GBMs.


Laboratory Investigation | 2018

Microvascular fractal dimension predicts prognosis and response to chemotherapy in glioblastoma: an automatic image analysis study

Cong Chen; Zhi cheng He; Yu Shi; Wenchao Zhou; Xia Zhang; Hua liang Xiao; Hai bo Wu; Xiao hong Yao; Wan chun Luo; You hong Cui; Shideng Bao; Hsiang-Fu Kung; Xiu Wu Bian; Yi Fang Ping

The microvascular profile has been included in the WHO glioma grading criteria. Nevertheless, microvessels in gliomas of the same WHO grade, e.g., WHO IV glioblastoma (GBM), exhibit heterogeneous and polymorphic morphology, whose possible clinical significance remains to be determined. In this study, we employed a fractal geometry-derived parameter, microvascular fractal dimension (mvFD), to quantify microvessel complexity and developed a home-made macro in Image J software to automatically determine mvFD from the microvessel-stained immunohistochemical images of GBM. We found that mvFD effectively quantified the morphological complexity of GBM microvasculature. Furthermore, high mvFD favored the survival of GBM patients as an independent prognostic indicator and predicted a better response to chemotherapy of GBM patients. When investigating the underlying relations between mvFD and tumor growth by deploying Ki67/mvFD as an index for microvasculature-normalized tumor proliferation, we discovered an inverse correlation between mvFD and Ki67/mvFD. Furthermore, mvFD inversely correlated with the expressions of a glycolytic marker, LDHA, which indicated poor prognosis of GBM patients. Conclusively, we developed an automatic approach for mvFD measurement, and demonstrated that mvFD could predict the prognosis and response to chemotherapy of GBM patients.


Journal of Cancer | 2018

Large Intergenic Non-coding RNA-RoR Inhibits Aerobic Glycolysis of Glioblastoma Cells via Akt Pathway

Yong Li; Zhi-Cheng He; Qing Liu; Kai Zhou; Yu Shi; Xiao-hong Yao; Xia Zhang; Hsiang-Fu Kung; Yi-Fang Ping; Xiu-wu Bian

Reprogramming energy metabolism is a hallmark of malignant tumors, including glioblastoma (GBM). Aerobic glycolysis is often utilized by tumor cells to maintain survival and proliferation. However, the underlying mechanisms of aerobic glycolysis in GBM remain elusive. Herein, we demonstrated that large intergenic non-coding RNA-RoR (LincRNA-RoR) functioned as a critical suppressor to inhibit the aerobic glycolysis and viability of GBM cells. We found that LincRNA-RoR was markedly reduced in GBM tissues compared with adjacent non-tumor tissues from 10 cases of GBM patients. Consistently, LincRNA-RoR expression in GBM cells was significantly lower than that in normal glial cells. The aerobic glycolysis of GBM cells, as determined by the measurement of glucose uptake and lactate production, was impaired by LincRNA-RoR overexpression. Mechanistically, LincRNA-RoR inhibited the expression of Rictor, the key component of mTORC2 (mammalian target of rapamycin complex 2), to suppress the activity of Akt pathway and impair the expression of glycolytic effectors, including Glut1, HK2, PKM2 and LDHA. Finally, enforced expression of LincRNA-RoR reduced the proliferation of GBM cells in vitro, restrained tumor growth in vivo, and repressed the expression of glycolytic molecules in GBM xenografts. Collectively, our results underscore LincRNA-RoR as a new suppressor of GBM aerobic glycolysis with therapeutic potential.


Cancer Letters | 2018

Stanniocalcin-1 augments stem-like traits of glioblastoma cells through binding and activating NOTCH1

Yong Li; Zhi-Cheng He; Xiao-Ning Zhang; Qing Liu; Cong Chen; Zheng Zhu; Qian Chen; Yu Shi; Xiao-hong Yao; You-Hong Cui; Xia Zhang; Yan Wang; Hsiang-Fu Kung; Yi-Fang Ping; Xiu-wu Bian

Glioblastoma (GBM) is a fatal tumor and comprises heterogeneous cells in which a subpopulation with stem cell-like properties is included. Cancer cells with stem cell-like properties account for tumor initiation, drug resistance and recurrence. To identify and characterize specific factors in regulating stem-like traits is critical for GBM therapeutic. Here, we showed that Stanniocalcin-1 (STC1), a secretory glycoprotein, functions as a novel stimulator for stem-like traits of GBM cells. We found STC1 was prominently expressed in glioma spheres which are mainly comprised of glioma stem-like cells. The stem-like traits of GBM cells, as determined by the expression of stem cell markers, tumor-sphere formation efficiency and colony-forming ability, were enhanced by STC1 overexpression and inhibited by STC1 knockdown. Furthermore, introduction of STC1 enhanced tumorigenesis inxa0vivo while knockdown of STC1 showed reverse effect. Finally, we demonstrated that STC1 interacted with the extracellular domain of NOTCH1 to activate NOTCH1-SOX2 signaling pathway, by which STC1 augmented the stem-like traits of GBM cells. Taken together, our data herein indicate that STC1 is a novel non-canonical NOTCH ligand and acts as a crucial regulator of stemness in GBM.


International Journal of Clinical and Experimental Pathology | 2014

Distinct patterns of ALDH1A1 expression predict metastasis and poor outcome of colorectal carcinoma

Sen-lin Xu; Dong-Zu Zeng; Weiguo Dong; Yanqing Ding; Jun Rao; Jiang-Jie Duan; Qing Liu; Jing Yang; Na Zhan; Ying Liu; Qi-Ping Hu; Xia Zhang; You-Hong Cui; Hsiang-Fu Kung; Shi-cang Yu; Xiu-wu Bian

Collaboration


Dive into the Hsiang-Fu Kung's collaboration.

Top Co-Authors

Avatar

Xiu-wu Bian

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xia Zhang

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yi-Fang Ping

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Xiao-hong Yao

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Qing Liu

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yong Lin

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Yu Shi

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Zhi-Cheng He

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

Cong Chen

Third Military Medical University

View shared research outputs
Top Co-Authors

Avatar

De-Yu Guo

Third Military Medical University

View shared research outputs
Researchain Logo
Decentralizing Knowledge