Network


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

Hotspot


Dive into the research topics where Jung-Tung Hung is active.

Publication


Featured researches published by Jung-Tung Hung.


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

Potent immune-modulating and anticancer effects of NKT cell stimulatory glycolipids

Ya-Jen Chang; Jing-Rong Huang; Yi-Chien Tsai; Jung-Tung Hung; Douglass Wu; Masakazu Fujio; Chi-Huey Wong; Alice L. Yu

α-Galactosylceramide (α-GalCer), a glycolipid that stimulates natural killer T (NKT) cells to produce both T helper (Th)1 and Th2 cytokines, has shown antitumor effects in mice but failed in clinical trials. We evaluated 16 analogs of α-GalCer for their CD1-mediated T cell receptor (TCR) activation of naïve human NKT cells and their anticancer efficacy. In vitro, glycolipids containing an aromatic ring in their acyl tail or sphingosine tail were more effective than α-GalCer in inducing Th1 cytokines/chemokines, TCR activation, and human NKT cell expansion. None of these glycolipids could directly stimulate human dendritic cell maturation, except for a glycolipid with an aromatic ring on the sphingosine tail. Here, we show that glycolipids activated the TCR of NKT cells with phosphorylation of CD3ε, ERK1/2, or CREB, which correlated with their induction of Th1 cytokines. Notably, the extent of NKT cell activation when glycolipid was presented by antigen-presenting cells was greater than when glycolipid was presented by non-antigen-presenting cells. In vivo, in mice bearing breast or lung cancers, the glycolipids that induced more Th1-biased cytokines and CD8/CD4 T cells displayed significantly greater anticancer potency than α-GalCer. These findings indicate that α-GalCer analogs can be designed to favor Th1-biased immunity, with greater anticancer efficacy and other immune-enhancing activities than α-GalCer itself.


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

Glycan microarray of Globo H and related structures for quantitative analysis of breast cancer

Cheng-Chi Wang; Yen-Lin Huang; Chien-Tai Ren; Chin-Wei Lin; Jung-Tung Hung; Jyh-Cherng Yu; Alice L. Yu; Chung-Yi Wu; Chi-Huey Wong

Cancer-associated carbohydrate antigens are often found on the surface of cancer cells. Understanding their roles in cancer progression will lead to the development of new therapeutics and high-sensitivity diagnostics for cancers. Globo H is a member of this family, which is highly expressed on breast cancer cells. Here, we report the development of a glycan microarray of Globo H and its analogs for measurement of the dissociation constants on surface (KD,surf) with three different monoclonal antibodies (VK-9, Mbr1, and anti-SSEA-3), to deduce their binding specificity. The glycan microarray was also used to detect the amount of antibodies present in the plasma of breast cancer patients and normal blood donors. It was shown that the amount of antibodies against Globo H from breast cancer patients were significantly higher than normal blood donors, providing a new tool for possible breast cancer diagnosis. Compared with the traditional ELISA method, this array method required only atto-mole amounts of materials and is more effective and more sensitive (5 orders of magnitude). The glycan microarray thus provides a new platform for use to monitor the immune response to carbohydrate epitopes after vaccine therapy or during the course of cancer progression.


Proceedings of the National Academy of Sciences of the United States of America | 2013

Carbohydrate-based vaccines with a glycolipid adjuvant for breast cancer

Yen-Lin Huang; Jung-Tung Hung; Sarah K.C. Cheung; Hsin-Yu Lee; Kuo-Ching Chu; Shiou-Ting Li; Yu-Chen Lin; Chien-Tai Ren; Ting-Jen R. Cheng; Tsui-Ling Hsu; Alice L. Yu; Chung-Yi Wu; Chi-Huey Wong

Globo H (GH) is a hexasaccharide specifically overexpressed on a variety of cancer cells and therefore, a good candidate for cancer vaccine development. To identify the optimal carrier and adjuvant combination, we chemically synthesized and linked GH to a carrier protein, including keyhole limpet hemocyanion, diphtheria toxoid cross-reactive material (CRM) 197 (DT), tetanus toxoid, and BSA, and combined with an adjuvant, and it was administered to mice for the study of immune response. Glycan microarray analysis of the antiserum obtained indicated that the combination of GH-DT adjuvanted with the α-galactosylceramide C34 has the highest enhancement of anti-GH IgG. Compared with the phase III clinical trial vaccine, GH–keyhole limpet hemocyanion/QS21, the GH-DT/C34 vaccine elicited more IgG antibodies, which are more selective for GH and the GH-related epitopes, stage-specific embryonic antigen 3 (SSEA3) and SSEA4, all of which were specifically overexpressed on breast cancer cells and breast cancer stem cells with SSEA4 at the highest level (>90%). We, therefore, further developed SSEA4-DT/C34 as a vaccine candidate, and after immunization, it was found that the elicited antibodies are also IgG-dominant and very specific for SSEA4.


Journal of Immunology | 2014

α-Galactosylceramide but Not Phenyl-Glycolipids Induced NKT Cell Anergy and IL-33–Mediated Myeloid-Derived Suppressor Cell Accumulation via Upregulation of egr2/3

Jing-Rong Huang; Yi-Chien Tsai; Ya-Jen Chang; Jen-Chien Wu; Jung-Tung Hung; Kun-Hsien Lin; Chi-Huey Wong; Alice L. Yu

Strategies for cancer immunotherapy include activating immune system for therapeutic benefit or blockade of immune checkpoints. To harness innate immunity to fight cancer, α-galactosylceramide (α-GalCer) has been used to activate NKT cells. Unfortunately, administration of α-GalCer causes long-term NKT cell anergy, but the molecular mechanism is unclear. In this study, we showed that α-GalCer–triggered egr2/3, which induced programmed death 1 and cbl-b in NKT cells, leading to NKT cell anergy. We also uncovered the induction of the immunosuppressive myeloid-derived suppressor cells (MDSCs) in the spleen by α-GalCer that might attenuate its antitumor efficacy. The accumulation of MDSC was accompanied by 20-fold rise in their arg-1 mRNAs and enhanced expression of programmed death 1/programmed death ligand 1. Furthermore, α-GalCer–induced egr-2/3 in hepatic NKT cells upregulated their TRAIL in addition to Fas ligand (FasL) and induced alarm signaling molecule IL-33 in Kupffer cells, presumably because of liver damage triggered by TRAIL/FasL. We further demonstrated that IL-33–stimulated macrophages produce G-CSF, which in turn, boosted MDSCs. Thus, α-GalCer–induced FasL/TRAIL and IL-33 provided a novel mechanism underlying α-GalCer–induced hepatotoxicity and MDSC accumulation. In contrast, analogs of α-GalCer containing phenyl group in the lipid tail could neither induce NKT anergy nor enhance MDSCs accumulation. Furthermore, tumor-infiltrating MDSCs in mice injected repeatedly with α-GalCer were 2-fold higher than those treated with phenyl-glycolipids. These results not only revealed the induction of MDSC via IL-33 as a new mechanism for α-GalCer–elicited immunosuppression but also provided one of the mechanisms underlying the superior antitumor potency of phenyl-glycolipids. Our findings have important implications for the development of NKT-stimulatory glycolipids as vaccine adjuvants and anticancer therapeutics.


Breast Cancer Research | 2014

Malignant phyllodes tumors display mesenchymal stem cell features and aldehyde dehydrogenase/disialoganglioside identify their tumor stem cells

Jin-Jin Lin; Chiun-Sheng Huang; John Yu; Guo-Shiou Liao; Huang-Chun Lien; Jung-Tung Hung; Ruey-Jen Lin; Kun-Tu Yeh; Alice L. Yu

IntroductionAlthough breast phyllodes tumors are rare, there is no effective therapy other than surgery. Little is known about their tumor biology. A malignant phyllodes tumor contains heterologous stromal elements, and can transform into rhabdomyosarcoma, liposarcoma and osteosarcoma. These versatile properties prompted us to explore their possible relationship to mesenchymal stem cells (MSCs) and to search for the presence of cancer stem cells (CSCs) in phyllodes tumors.MethodsParaffin sections of malignant phyllodes tumors were examined for various markers by immunohistochemical staining. Xenografts of human primary phyllodes tumors were established by injecting freshly isolated tumor cells into the mammary fat pad of non-obese diabetic-severe combined immunodeficient (NOD-SCID) mice. To search for CSCs, xenografted tumor cells were sorted into various subpopulations by flow cytometry and examined for their in vitro mammosphere forming capacity, in vivo tumorigenicity in NOD-SCID mice and their ability to undergo differentiation.ResultsImmunohistochemical analysis revealed the expression of the following 10 markers: CD44, CD29, CD106, CD166, CD105, CD90, disialoganglioside (GD2), CD117, Aldehyde dehydrogenase 1 (ALDH), and Oct-4, and 7 clinically relevant markers (CD10, CD34, p53, p63, Ki-67, Bcl-2, vimentin, and Globo H) in all 51 malignant phyllodes tumors examined, albeit to different extents. Four xenografts were successfully established from human primary phyllodes tumors. In vitro, ALDH+ cells sorted from xenografts displayed approximately 10-fold greater mammosphere-forming capacity than ALDH- cells. GD2+ cells showed a 3.9-fold greater capacity than GD2- cells. ALDH+/GD2+cells displayed 12.8-fold greater mammosphere forming ability than ALDH-/GD2- cells. In vivo, the tumor-initiating frequency of ALDH+/GD2+ cells were up to 33-fold higher than that of ALDH+ cells, with as few as 50 ALDH+/GD2+ cells being sufficient for engraftment. Moreover, we provided the first evidence for the induction of ALDH+/GD2+ cells to differentiate into neural cells of various lineages, along with the observation of neural differentiation in clinical specimens and xenografts of malignant phyllodes tumors. ALDH+ or ALDH+/GD2+ cells could also be induced to differentiate into adipocytes, osteocytes or chondrocytes.ConclusionsOur findings revealed that malignant phyllodes tumors possessed many characteristics of MSC, and their CSCs were enriched in ALDH+ and ALDH+/GD2+ subpopulations.


Cancer Research | 2014

Globo-H Ceramide Shed from Cancer Cells Triggers Translin-Associated Factor X-Dependent Angiogenesis

Jing-Yan Cheng; Sheng-Hung Wang; Juway Lin; Yi-Chien Tsai; John Yu; Jen-Chine Wu; Jung-Tung Hung; Jin-Jin Lin; Yih-Yiing Wu; Kun-Tu Yeh; Alice L. Yu

Tumor angiogenesis is a critical element of cancer progression, and strategies for its selective blockade are still sought. Here, we examine the angiogenic effects of Globo-H ceramide (GHCer), the most prevalent glycolipid in a majority of epithelial cancers and one that acts as an immune checkpoint. Here, we report that GHCer becomes incorporated into endothelial cells through the absorption of microvesicles shed from tumor cells. In endothelial cells, GHCer addition induces migration, tube formation, and intracellular Ca(2+) mobilization in vitro and angiogenesis in vivo. Breast cancer cells expressing high levels of GHCer displayed relatively greater tumorigenicity and angiogenesis compared with cells expressing low levels of Globo-H. Clincally, GHCer(+) breast cancer specimens contained higher vessel density than GHCer(-) breast cancer specimens. Mechanistic investigations linked the angiogenic effects of GHCer to its endocytosis and binding to TRAX, with consequent release of PLCβ1 from TRAX to trigger Ca(2+) mobilization. Together, our findings highlight the importance of GHC as a target for cancer therapy by providing new information on its key role in tumor angiogenesis.


Cell Death and Disease | 2016

Fucosylation of LAMP-1 and LAMP-2 by FUT1 correlates with lysosomal positioning and autophagic flux of breast cancer cells

Keng-Poo Tan; Ming-Yi Ho; Huan-Chieh Cho; John Yu; Jung-Tung Hung; Alice L. Yu

Alpha1,2-fucosyltransferases, FUT1 and FUT2, which transfer fucoses onto the terminal galactose of N-acetyl-lactosamine via α1,2-linkage have been shown to be highly expressed in various types of cancers. A few studies have shown the involvement of FUT1 substrates in tumor cell proliferation and migration. Lysosome-associated membrane protein 1, LAMP-1, has been reported to carry alpha1,2-fucosylated Lewis Y (LeY) antigens in breast cancer cells, however, the biological functions of LeY on LAMP-1 remain largely unknown. Whether or not its family member, LAMP-2, displays similar modifications and functions as LAMP-1 has not yet been addressed. In this study, we have presented evidence supporting that both LAMP-1 and 2 are substrates for FUT1, but not FUT2. We have also demonstrated the presence of H2 and LeY antigens on LAMP-1 by a targeted nanoLC-MS3 and the decreased levels of fucosylation on LAMP-2 by MALDI-TOF analysis upon FUT1 knockdown. In addition, we found that the expression of LeY was substantial in less invasive ER+/PR+/HER− breast cancer cells (MCF-7 and T47D) but negligible in highly invasive triple-negative MDA-MB-231 cells, of which LeY levels were correlated with the levels of LeY carried by LAMP-1 and 2. Intriguingly, we also observed a striking change in the subcellular localization of lysosomes upon FUT1 knockdown from peripheral distribution of LAMP-1 and 2 to a preferential perinuclear accumulation. Besides that, knockdown of FUT1 led to an increased rate of autophagic flux along with diminished activity of mammalian target of rapamycin complex 1 (mTORC1) and enhanced autophagosome–lysosome fusion. This may be associated with the predominantly perinuclear distribution of lysosomes mediated by FUT1 knockdown as lysosomal positioning has been reported to regulate mTOR activity and autophagy. Taken together, our results suggest that downregulation of FUT1, which leads to the perinuclear localization of LAMP-1 and 2, is correlated with increased rate of autophagic flux by decreasing mTOR signaling and increasing autolysosome formation.


Journal of Cancer Science & Therapy | 2013

A Prevalent Cancer Associated Glycan, Globo H Ceramide, Induces Immunosuppression by Reducing Notch1 Signaling

Yi-Chien Tsai; Jing-Rong Huang; Jing-Yan Cheng; Jin-Jin Lin; Jung-Tung Hung; Yih-Yiing Wu; Kun-Tu Yeh; Alice L. Yu

Globo H, a hexasaccharide initially identified as a ceramide-linked glycolipid from human breast cancer cell line MCF-7, is over-expressed on the surface of many common cancers, but its function is unknown. Here we demonstrated the uptake of globo H ceramide (GHCer) by human peripheral blood mononuclear cells (PBMC) upon co-culture with MCF-7 cells. Significantly, the expression of globo H on tumor-infiltrating lymphocytes was observed in 61% of globo H positive breast cancer tissues. Addition of synthetic GHCer to human PBMC, mouse splenocytes or purified CD4+ T cells inhibited their proliferative response to anti-CD3/CD28 to 60 ± 1%, 50 ± 7% and 62 ± 5% of control, respectively, and reduced the secretion of IL-2, IFN-γ and IL-4. GHCer also significantly suppressed the proliferation of splenocytes or purified CD19+ B cells to 45 ± 10% and 26 ± 3% of control in response to LPS or LPS + IL4 +CD40 ligand and their IgM production to 12 ± 5% and, 8 ± 3%, and IgG to 34 ± 9%, 35 ± 5%, respectively, with neglible induction of plasma cells. Ceramide displayed no such inhibitory effects. On the other hand, GHCer failed to raise the number of regulatory T cells, or their expression of FOXP3/CTLA4, nor did it increase apoptosis. Notably, GHCer significantly suppressed the Notch1 signaling during activation of human PBMC and murine T and B cells. Furthermore, GHCer upregulated the expression of ID3 and itch by 2.1±0.2 and 4.7 ± 0.4 fold, respectively, leading to ID3-dependent downregulation of Notch1 and itch-mediated Notch1 degradation. These results provide the first evidence for GHCer to facilitate the escape of cancer cells from immune surveillance.


ChemBioChem | 2012

Synthesis and Evaluation of Acyl-Chain- and Galactose-6′′-Modified Analogues of α-GalCer for NKT Cell Activation

Ming‐Han Hsieh; Jung-Tung Hung; Ya‐Wen Liw; Yin‐Jen Lu; Chi-Huey Wong; Alice L. Yu; Pi-Hui Liang

α‐GalCer is an immunostimulating glycolipid that binds to CD1d molecules and activates invariant natural killer T (iNKT) cells. Here we report a scaled‐up synthesis of α‐GalCer analogues with modifications in the acyl side chain and/or at the galactose 6′′‐position, together with their evaluation in vitro and in vivo. Analogues containing 11‐phenylundecanoyl acyl side chains with aromatic substitutions (14, 16–21) and Gal‐6′′‐phenylacetamide‐substituted α‐GalCer analogues bearing p‐nitro‐ (32), p‐tert‐butyl (34), or o‐, m‐, or p‐methyl groups (40–42) displayed higher IFN‐γ/IL‐4 secretion ratios than α‐GalCer in vitro. In mice, compound 16, with an 11‐(3,4‐difluorophenyl)undecanoyl acyl chain, induced significant proliferation of NK and DC cells, which should be beneficial in killing tumors and priming the immune response. These new glycolipids might prove useful as adjuvants or anticancer agents.


Antiviral Research | 2014

Potent adjuvant effects of novel NKT stimulatory glycolipids on hemagglutinin based DNA vaccine for H5N1 influenza virus.

Jung-Tung Hung; Yi-Chieh Tsai; Wen-Der Lin; Jia-Tsrong Jan; Kun-Hsien Lin; Jing-Rong Huang; Jing-Yan Cheng; Ming-Wei Chen; Chi-Huey Wong; Alice L. Yu

H5N1 influenza virus is a highly pathogenic virus, posing a pandemic threat. Previously, we showed that phenyl analogs of α-galactosylceramide (α-GalCer) displayed greater NKT stimulation than α-GalCer. Here, we examined the adjuvant effects of one of the most potent analogs, C34, on consensus hemagglutinin based DNA vaccine (pCHA5) for H5N1 virus. Upon intramuscular electroporation of mice with pCHA5 with/without various α-GalCer analogs, C34-adjuvanted group developed the highest titer against consensus H5 and more HA-specific IFN-γ secreting CD8 cells (203±13.5) than pCHA5 alone (152.6±13.7, p<0.05). Upon lethal challenge of NIBRG-14 virus, C34-adjuvanted group (84.6%) displayed higher survival rate than pCHA5 only group (46.1%). In the presence of C34 as adjuvant, the antisera displayed broader and greater neutralizing activities against virions pseudotyped with HA of clade 1, and 2.2 than pCHA5 only group. Moreover, to simulate an emergency response to a sudden H5N1 outbreak, we injected mice intramuscularly with single dose of a new consensus H5 (pCHA5-II) based on 1192 full-length H5 sequences, with C34 as adjuvant. The latter not only enhanced the humoral immune response and protection against virus challenge, but also broadened the spectrum of neutralization against pseudotyped HA viruses. Our vaccine strategy can be easily implemented for any H5N1 virus outbreak by single IM injection of a consensus H5 DNA vaccine based on updated HA sequences using C34 as an adjuvant.

Collaboration


Dive into the Jung-Tung Hung's collaboration.

Top Co-Authors

Avatar

Alice L. Yu

University of California

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

John Yu

Memorial Hospital of South Bend

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Top Co-Authors

Avatar

Sheng-Hung Wang

Memorial Hospital of South Bend

View shared research outputs
Top Co-Authors

Avatar
Top Co-Authors

Avatar
Researchain Logo
Decentralizing Knowledge