Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters











Publication year range
1.
Biomed Pharmacother ; 177: 117040, 2024 Aug.
Article in English | MEDLINE | ID: mdl-38959605

ABSTRACT

Invariant natural killer T cell (iNKT) cells produce large amounts of cytokines in response to α-Galactosylceramide (α-GalCer) stimulation. An analog containing two phenyl rings on the acyl chain, C34, was previously found to be more Th1-biased than α-GalCer and triggered greater anticancer activities against breast cancer, melanoma and lung cancer in mice. Since liver is enriched in iNKT cells, we investigated anticancer efficacy of C34 on neuroblastoma with hepatic metastasis. C34 induced Th1-biased cytokine secretions in the liver, significantly suppressed neuroblastoma growth/metastasis and prolonged mouse survival. The anti-tumor efficacy might be attributed to greater expansions of hepatic NKT, NK, CD4+ T, and CD8+ T cells as well as reduction of the number of SSCloGr1intCD11b+ subset of myeloid-derived suppressor cells (MDSCs) in the liver of tumor-bearing mice, as compared to DMSO control group. C34 also upregulated expression of CD1d and CD11c, especially in the SSCloGr1intCD11b+ subset of MDSCs, which might be killed by C34-activated NKT cells, attributing to their reduced number. In addition, C34 also induced expansion of CD4+ T, CD8+ T, and NK cells, which might eliminate neuroblastoma cells. These immune-modulating effects of C34 might act in concert in the local milieu of liver to suppress the tumor growth. Further analysis of database of neuroblastoma revealed that patients with high CD11c expression in the monocytic MDSCs in the tumor had longer survival, suggesting the potential clinical application of C34 for treatment of neuroblastoma.


Subject(s)
Glycolipids , Liver Neoplasms , Natural Killer T-Cells , Neuroblastoma , Animals , Neuroblastoma/pathology , Neuroblastoma/drug therapy , Neuroblastoma/immunology , Natural Killer T-Cells/drug effects , Natural Killer T-Cells/immunology , Natural Killer T-Cells/metabolism , Liver Neoplasms/drug therapy , Liver Neoplasms/pathology , Liver Neoplasms/immunology , Liver Neoplasms/secondary , Cell Line, Tumor , Mice , Glycolipids/pharmacology , Humans , Female , Cytokines/metabolism , Myeloid-Derived Suppressor Cells/drug effects , Myeloid-Derived Suppressor Cells/immunology , Myeloid-Derived Suppressor Cells/metabolism , Antineoplastic Agents/pharmacology , Galactosylceramides/pharmacology
2.
Oncogene ; 40(16): 2858-2871, 2021 04.
Article in English | MEDLINE | ID: mdl-33742122

ABSTRACT

Cancer stem cells (CSC) play a pivotal role in cancer metastasis and resistance to therapy. Previously, we compared the phosphoproteomes of breast cancer stem cells (BCSCs) enriched subpopulation and non-BCSCs sorted from breast cancer patient-derived xenograft (PDX), and identified a function unknown protein, transmembrane and coiled-coil domain family 3 (TMCC3) to be a potential enrichment marker for BCSCs. We demonstrated greater expression of TMCC3 in BCSCs than non-BCSCs and higher expression of TMCC3 in metastatic lymph nodes and lungs than in primary tumor of breast cancer PDXs. TMCC3 silencing suppressed mammosphere formation, ALDH activity and cell migration in vitro, along with reduced tumorigenicity and metastasis in vivo. Mechanistically, we found that AKT activation was reduced by TMCC3 silencing, but enhanced by TMCC3 overexpression. We further demonstrated that TMCC3 interacted directly with AKT through its 1-153 a.a. domain by cell-free biochemical assay in vitro and co-immunoprecipitation and interaction domain mapping assays in vivo. Based on domain truncation studies, we showed that the AKT-interacting domain of TMCC3 was essential for TMCC3-induced AKT activation, self-renewal, and metastasis. Clinically, TMCC3 mRNA expression in 202 breast cancer specimens as determined by qRT-PCR assay showed that higher TMCC3 expression correlated with poorer clinical outcome of breast cancer, including early-stage breast cancer. Multivariable analysis identified TMCC3 expression as an independent risk factor for survival. These findings suggest that TMCC3 is crucial for maintenance of BCSCs features through AKT regulation, and TMCC3 expression has independent prognostic significance in breast cancer. Thus, TMCC3 may serve as a new target for therapy directed against CSCs.


Subject(s)
Breast Neoplasms/metabolism , Membrane Proteins/metabolism , Neoplastic Stem Cells/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Animals , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Female , Heterografts , Humans , Membrane Proteins/genetics , Mice , Oncogenes , Proto-Oncogene Proteins c-akt/genetics , Risk Factors
3.
Front Immunol ; 12: 791551, 2021.
Article in English | MEDLINE | ID: mdl-35046949

ABSTRACT

Synopsis: A sugar-lipid molecule called OAcGD2 is a novel marker for breast cancer stem cells. Treatment with anti-OAcGD2 mAb8B6 may have superior anticancer efficacy by targeting cancer stem cells, thereby reducing metastasis and recurrence of cancer. Background: Cancer stem cells (CSCs) that drive tumor progression and disease recurrence are rare subsets of tumor cells. CSCs are relatively resistant to conventional chemotherapy and radiotherapy. Eradication of CSCs is thus essential to achieve durable responses. GD2 was reported to be a CSC marker in human triple-negative breast cancer, and anti-GD2 immunotherapy showed reduced tumor growth in cell lines. Using a specific anti-OAcGD2 antibody, mAb8D6, we set out to determine whether OAcGD2+ cells exhibit stem cell properties and mAb8D6 can inhibit tumor growth by targeting OAcGD2+CSCs. Method: OAcGD2 expression in patient-derived xenografts (PDXs) of breast cancer was determined by flow cytometric analyses using mAb8D6. The stemness of OAcGD2+ cells isolated by sorting and the effects of mAb8B6 were assessed by CSC growth and mammosphere formation in vitro and tumor growth in vivo using PDX models. Result: We found that the OAcGD2 expression levels in six PDXs of various molecular subtypes of breast cancer highly correlated with their previously defined CSC markers in these PDXs. The sorted OAcGD2+ cells displayed a greater capacity for mammosphere formation in vitro and tumor initiation in vivo than OAcGD2- cells. In addition, the majority of OAcGD2+ cells were aldehyde dehydrogenase (ALDH+) or CD44hiCD24lo, the known CSC markers in breast cancer. Treatment of PDXs-bearing mice with mAb8B6, but not doxorubicin, suppressed the tumor growth, along with reduced CSCs as assessed by CSC markers and in vivo tumorigenicity. In vitro, mAb8B6 suppressed proliferation and mammosphere formation and induced apoptosis of OAcGD2+ breast cancer cells harvested from PDXs, in a dose-dependent manner. Finally, administration of mAb8B6 in vivo dramatically suppressed tumor growth of OAcGD2+ breast CSCs (BCSCs) with complete tumor abrogation in 3/6 mice. Conclusion: OAcGD2 is a novel marker for CSC in various subtypes of breast cancer. Anti-OAcGD2 mAb8B6 directly eradicated OAcGD2+ cells and reduced tumor growth in PDX model. Our data demonstrate the potential of mAb8B6 as a promising immunotherapeutic agent to target BCSCs.


Subject(s)
Antibodies, Monoclonal/pharmacology , Breast Neoplasms/pathology , Gangliosides/metabolism , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Animals , Apoptosis/drug effects , Biomarkers , Cell Proliferation/drug effects , Female , Humans , Mice , Xenograft Model Antitumor Assays
4.
Chem Commun (Camb) ; 56(43): 5827-5830, 2020 May 28.
Article in English | MEDLINE | ID: mdl-32329494

ABSTRACT

Thrombin activates protease-activated receptor-1 (PAR-1) through binding to exosite I and the active site to promote tumor growth. We have developed a new class of anti-cancer glyco-peptides to target exosite I selectively without affecting the active-site-mediated coagulation activity and showed the importance of glycans for the stability and anti-cancer activity of the glyco-peptides.


Subject(s)
Antineoplastic Agents/therapeutic use , Glycopeptides/therapeutic use , Neoplasms/drug therapy , Receptor, PAR-1/metabolism , Thrombin/metabolism , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Line, Tumor , Drug Design , Glycopeptides/chemistry , Glycopeptides/pharmacology , Humans , Mice, SCID , Neoplasms/metabolism , Neoplasms/pathology , Thrombin/chemistry
5.
Int J Cancer ; 146(6): 1674-1685, 2020 03 15.
Article in English | MEDLINE | ID: mdl-31340060

ABSTRACT

G protein-coupled estrogen receptor-1 (GPER), a member of the G protein-coupled receptor (GPCR) superfamily, mediates estrogen-induced proliferation of normal and malignant breast epithelial cells. However, its role in breast cancer stem cells (BCSCs) remains unclear. Here we showed greater expression of GPER in BCSCs than non-BCSCs of three patient-derived xenografts of ER- /PR+ breast cancers. GPER silencing reduced stemness features of BCSCs as reflected by reduced mammosphere forming capacity in vitro, and tumor growth in vivo with decreased BCSC populations. Comparative phosphoproteomics revealed greater GPER-mediated PKA/BAD signaling in BCSCs. Activation of GPER by its ligands, including tamoxifen (TMX), induced phosphorylation of PKA and BAD-Ser118 to sustain BCSC characteristics. Transfection with a dominant-negative mutant BAD (Ser118Ala) led to reduced cell survival. Taken together, GPER and its downstream signaling play a key role in maintaining the stemness of BCSCs, suggesting that GPER is a potential therapeutic target for eradicating BCSCs.


Subject(s)
Breast Neoplasms/pathology , Neoplastic Stem Cells/pathology , Receptors, Estrogen/metabolism , Receptors, G-Protein-Coupled/metabolism , Animals , Breast/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Cyclic AMP-Dependent Protein Kinases/metabolism , Female , Gene Knockdown Techniques , Humans , Mice , Phosphorylation/drug effects , Receptors, Estrogen/genetics , Receptors, G-Protein-Coupled/agonists , Receptors, G-Protein-Coupled/genetics , Receptors, Progesterone/metabolism , Signal Transduction/drug effects , Spheroids, Cellular , Tamoxifen/pharmacology , Xenograft Model Antitumor Assays , bcl-Associated Death Protein/metabolism
6.
EBioMedicine ; 45: 25-38, 2019 Jul.
Article in English | MEDLINE | ID: mdl-31262713

ABSTRACT

BACKGROUND: The transcription factor Nrf2 is a master regulator of antioxidant response. While Nrf2 activation may counter increasing oxidative stress in aging, its activation in cancer can promote cancer progression and metastasis, and confer resistance to chemotherapy and radiotherapy. Thus, Nrf2 has been considered as a key pharmacological target. Unfortunately, there are no specific Nrf2 inhibitors for therapeutic application. Moreover, high Nrf2 activity in many tumors without Keap1 or Nrf2 mutations suggests that alternative mechanisms of Nrf2 regulation exist. METHODS: Interaction of FAM129B with Keap1 is demonstrated by immunofluorescence, colocalization, co-immunoprecipitation and mammalian two-hybrid assay. Antioxidative function of FAM129B is analyzed by measuring ROS levels with DCF/flow cytometry, Nrf2 activation using luciferase reporter assay and determination of downstream gene expression by qPCR and wester blotting. Impact of FAM129B on in vivo chemosensitivity is examined in mice bearing breast and colon cancer xenografts. The clinical relevance of FAM129B is assessed by qPCR in breast cancer samples and data mining of publicly available databases. FINDINGS: We have demonstrated that FAM129B in cancer promotes Nrf2 activity by reducing its ubiquitination through competition with Nrf2 for Keap1 binding via its DLG and ETGE motifs. In addition, FAM129B reduces chemosensitivity by augmenting Nrf2 antioxidative signaling and confers poor prognosis in breast and lung cancer. INTERPRETATION: These findings demonstrate the important role of FAM129B in Nrf2 activation and antioxidative response, and identify FMA129B as a potential therapeutic target. FUND: The Chang Gung Medical Foundation (Taiwan) and the Ministry of Science and Technology (Taiwan).


Subject(s)
Antioxidants/metabolism , Kelch-Like ECH-Associated Protein 1/genetics , NF-E2-Related Factor 2/genetics , Oxidative Stress/genetics , Phosphoproteins/genetics , Aging/genetics , Animals , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Colonic Neoplasms/drug therapy , Colonic Neoplasms/genetics , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Regulation, Neoplastic , HCT116 Cells , Humans , Mice , Protein Binding/genetics , Xenograft Model Antitumor Assays
7.
Cell Death Discov ; 5: 74, 2019.
Article in English | MEDLINE | ID: mdl-30854233

ABSTRACT

FUT1 and FUT2 encode alpha 1, 2-fucosyltransferases which catalyze the addition of alpha 1, 2-linked fucose to glycans. Glycan products of FUT1 and FUT2, such as Globo H and Lewis Y, are highly expressed on malignant tissues, including breast cancer. Herein, we investigated the roles of FUT1 and FUT2 in breast cancer. Silencing of FUT1 or FUT2 by shRNAs inhibited cell proliferation in vitro and tumorigenicity in mice. This was associated with diminished properties of cancer stem cell (CSC), including mammosphere formation and CSC marker both in vitro and in xenografts. Silencing of FUT2, but not FUT1, significantly changed the cuboidal morphology to dense clusters of small and round cells with reduced adhesion to polystyrene and extracellular matrix, including laminin, fibronectin and collagen. Silencing of FUT1 or FUT2 suppressed cell migration in wound healing assay, whereas FUT1 and FUT2 overexpression increased cell migration and invasion in vitro and metastasis of breast cancer in vivo. A decrease in mesenchymal like markers such as fibronectin, vimentin, and twist, along with increased epithelial like marker, E-cadherin, was observed upon FUT1/2 knockdown, while the opposite was noted by overexpression of FUT1 or FUT2. As expected, FUT1 or FUT2 knockdown reduced Globo H, whereas FUT1 or FUT2 overexpression showed contrary effects. Exogenous addition of Globo H-ceramide reversed the suppression of cell migration by FUT1 knockdown but not the inhibition of cell adhesion by FUT2 silencing, suggesting that at least part of the effects of FUT1/2 knockdown were mediated by Globo H. Our results imply that FUT1 and FUT2 play important roles in regulating growth, adhesion, migration and CSC properties of breast cancer, and may serve as therapeutic targets for breast cancer.

8.
Int J Cancer ; 144(8): 1996-2007, 2019 04 15.
Article in English | MEDLINE | ID: mdl-30252131

ABSTRACT

ST3Gal1 is a key sialyltransferase which adds α2,3-linked sialic acid to substrates and generates core 1 O-glycan structure. Upregulation of ST3Gal1 has been associated with worse prognosis of breast cancer patients. However, the protein substrates of ST3Gal1 implicated in tumor progression remain elusive. In our study, we demonstrated that ST3GAL1-silencing significantly reduced tumor growth along with a notable decrease in vascularity of MCF7 xenograft tumors. We identified vasorin (VASN) which was shown to bind TGF-ß1, as a potential candidate that links ST3Gal1 to angiogenesis. LC-MS/MS analysis of VASN secreted from MCF7, revealed that more than 80% of its O-glycans are sialyl-3T and disialyl-T. ST3GAL1-silencing or desialylation of VASN by neuraminidase enhanced its binding to TGF-ß1 by 2- to 3-fold and thereby dampening TGF-ß1 signaling and angiogenesis, as indicated by impaired tube formation of HUVECs, suppressed angiogenesis gene expression and reduced activation of Smad2 and Smad3 in HUVEC cells. Examination of 114 fresh primary breast cancer and their adjacent normal tissues showed that the expression levels of ST3Gal1 and TGFB1 were high in tumor part and the expression of two genes was positively correlated. Kaplan Meier survival analysis showed a significantly shorter relapse-free survival for those with lower expression VASN, notably, the combination of low VASN with high ST3GAL1 yielded even higher risk of recurrence (p = 0.025, HR = 2.967, 95% CI = 1.14-7.67). Since TGF-ß1 is known to transcriptionally activate ST3Gal1, our findings illustrated a feedback regulatory loop in which TGF-ß1 upregulates ST3Gal1 to circumvent the negative impact of VASN.


Subject(s)
Breast Neoplasms/pathology , Carrier Proteins/metabolism , Membrane Proteins/metabolism , Neoplasm Recurrence, Local/pathology , Neovascularization, Pathologic/pathology , Sialyltransferases/metabolism , Transforming Growth Factor beta1/metabolism , Animals , Breast/pathology , Breast Neoplasms/blood supply , Breast Neoplasms/mortality , Disease Progression , Female , Human Umbilical Vein Endothelial Cells , Humans , MCF-7 Cells , Mice , Neoplasm Recurrence, Local/epidemiology , RNA, Small Interfering/metabolism , Sialic Acids/metabolism , Sialyltransferases/genetics , Signal Transduction , Survival Analysis , Up-Regulation , Xenograft Model Antitumor Assays , beta-Galactoside alpha-2,3-Sialyltransferase
9.
ACS Chem Biol ; 11(12): 3431-3441, 2016 12 16.
Article in English | MEDLINE | ID: mdl-27782396

ABSTRACT

Glycosphingolipids (GSLs) bearing the α-galactosyl headgroup and the acyl chain terminated with a phenyl derivative were found to be more potent than α-galactosyl ceramide (αGalCer) to stimulate both murine and human invariant natural killer T (iNKT) cells and to induce an antibody isotope switch to IgG. In this study, we replaced the galactosyl group with glucose (αGlc) and its fluoro-analogs and found that phenyl GSLs with αGlc (C34-Glc) and its fluoro-analog 6F-C34-Glc were stronger than those with αGal in stimulating human iNKT cells but weaker in mice. Their activities have a strong correlation with the binding avidities of the ternary interaction between the iNKT-cell receptor (iNKTCR) and CD1d-GSL complex. It was the iNKTCR rather than CD1d that dictated the species-specific responses. C34-Glc was further used as an adjuvant for a SSEA4-crm-197 vaccine, and after immunization in mice, the vaccine was highly effective against Lewis lung carcinoma.


Subject(s)
Adjuvants, Immunologic/chemistry , Adjuvants, Immunologic/pharmacology , Glycolipids/chemistry , Glycolipids/pharmacology , Lymphocyte Activation/drug effects , Natural Killer T-Cells/drug effects , Animals , Cancer Vaccines/chemistry , Cancer Vaccines/pharmacology , Cell Line , Humans , Mice , Mice, Inbred C57BL , Models, Molecular , Natural Killer T-Cells/immunology
10.
PLoS One ; 10(7): e0132579, 2015.
Article in English | MEDLINE | ID: mdl-26207989

ABSTRACT

DNA topoisomerase I (TOP1) levels of several human neoplasms are higher than those of normal tissues. TOP1 inhibitors are widely used in treating conventional therapy-resistant ovarian cancers. However, patients may develop resistance to TOP1 inhibitors, hampering chemotherapy success. In this study, we examined the mechanisms associated with the development of camptothecin (CPT) resistance in ovarian cancers and identified evodiamine (EVO), a natural product with TOP1 inhibiting activity that overcomes the resistance. The correlations among TOP1 levels, cancer staging, and overall survival (OS) were analyzed. The effect of EVO on CPT-resistant ovarian cancer was evaluated in vitro and in vivo. TOP1 was associated with poor prognosis in ovarian cancers (p = 0.024). EVO induced apoptosis that was detected using flow cytometry and terminal deoxynucleotidyl transferase dUTP nick end labeling (TUNEL) assay. The tumor size decreased significantly in the EVO treatment group compared with the control group (p < 0.01) in a xenograft mouse model. Effects of drugs targeting TOP1 for prognosis and therapy in CPT-resistant ovarian cancer are anticipated. EVO with TOP1 can be developed as an antiproliferative agent for overcoming CPT resistance in ovarian cancers.


Subject(s)
DNA Topoisomerases, Type I/metabolism , Drug Resistance, Neoplasm/drug effects , Indole Alkaloids/administration & dosage , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Animals , Antineoplastic Agents, Phytogenic/pharmacology , Apoptosis , Camptothecin/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Female , Humans , Indole Alkaloids/pharmacology , Mice , Ovarian Neoplasms/enzymology , Treatment Outcome , Xenograft Model Antitumor Assays
11.
PLoS One ; 10(7): e0134110, 2015.
Article in English | MEDLINE | ID: mdl-26218133

ABSTRACT

Chemo- and radiotherapy cause multiple forms of DNA damage and lead to the death of cancer cells. Inhibitors of the DNA damage response are candidate drugs for use in combination therapies to increase the efficacy of such treatments. In this study, we show that curcumin, a plant polyphenol, sensitizes budding yeast to DNA damage by counteracting the DNA damage response. Following DNA damage, the Mec1-dependent DNA damage checkpoint is inactivated and Rad52 recombinase is degraded by curcumin, which results in deficiencies in double-stand break repair. Additive effects on damage-induced apoptosis and the inhibition of damage-induced autophagy by curcumin were observed. Moreover, rpd3 mutants were found to mimic the curcumin-induced suppression of the DNA damage response. In contrast, hat1 mutants were resistant to DNA damage, and Rad52 degradation was impaired following curcumin treatment. These results indicate that the histone deacetylase inhibitor activity of curcumin is critical to DSB repair and DNA damage sensitivity.


Subject(s)
Curcumin/pharmacology , DNA Damage/drug effects , DNA Repair/drug effects , Drug Resistance/drug effects , Histone Deacetylases/chemistry , Saccharomyces cerevisiae/drug effects , Saccharomyces cerevisiae/metabolism , Acetylation/drug effects , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Cycle Checkpoints/drug effects , Cell Proliferation/drug effects , Chromatin Immunoprecipitation , Histone Deacetylase Inhibitors/pharmacology , Immunoblotting , Saccharomyces cerevisiae/genetics , Signal Transduction/drug effects
12.
Cancer Res ; 74(23): 6856-66, 2014 Dec 01.
Article in English | MEDLINE | ID: mdl-25281721

ABSTRACT

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 PLCß1 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.


Subject(s)
Antigens, Tumor-Associated, Carbohydrate/metabolism , Breast Neoplasms/blood supply , Ceramides/metabolism , DNA-Binding Proteins/metabolism , Animals , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Calcium/metabolism , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line , Cell Line, Tumor , Cell Movement/physiology , Cytoplasmic Vesicles/metabolism , Cytoplasmic Vesicles/pathology , Endocytosis/physiology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Female , Human Umbilical Vein Endothelial Cells , Humans , MCF-7 Cells , Mice , Mice, Inbred BALB C , Mice, Inbred NOD , Mice, SCID , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology
13.
Diabetes ; 57(7): 1861-9, 2008 Jul.
Article in English | MEDLINE | ID: mdl-18420489

ABSTRACT

OBJECTIVE: Coinhibitory signals mediated via programmed death 1 (PD-1) receptor play a critical role in downregulating immune responses and in maintaining peripheral tolerance. Programmed death 1 ligand 1 (PD-L1), the interacting ligand for PD-1, widely expressed in many cell types, acts as a tissue-specific negative regulator of pathogenic T-cell responses. We investigated the protective potential of PD-L1 on autoimmune diabetes by transgenically overexpressing PD-L1 in pancreatic beta-cells in nonobese diabetic (NOD) mice. RESEARCH DESIGN AND METHODS: We established an insulin promoter-driven murine PD-L1 transgenic NOD mouse model to directly evaluate the protective effect of an organ-specific PD-L1 transgene against autoimmune diabetes. Transgene expression, insulitis, and diabetic incidence were characterized in these transgenic NOD mice. Lymphocyte development, Th1 cells, and regulatory T-cells were analyzed in these transgenic mice; and T-cell proliferation, adoptive transfer, and islet transplantation were performed to evaluate the PD-L1 transgene-mediated immune-protective mechanisms. RESULTS: The severity of insulitis in these transgenic mice is significantly decreased, disease onset is delayed, and the incidence of diabetes is markedly decreased compared with littermate controls. NOD/SCID mice that received lymphocytes from transgenic mice became diabetic at a slower rate than mice receiving control lymphocytes. Moreover, lymphocytes collected from recipients transferred by lymphocytes from transgenic mice revealed less proliferative potential than lymphocytes obtained from control recipients. Transgenic islets transplanted in diabetic recipients survived moderately longer than control islets. CONCLUSIONS: Our results demonstrate the protective potential of transgenic PD-L1 in autoimmune diabetes and illustrate its role in downregulating diabetogenic T-cells in NOD mice.


Subject(s)
B7-1 Antigen/genetics , Diabetes Mellitus, Type 1/immunology , Membrane Glycoproteins/genetics , Peptides/genetics , Adoptive Transfer , Animals , B7-1 Antigen/analysis , B7-1 Antigen/physiology , B7-H1 Antigen , Cell Division , Cloning, Molecular , Diabetes Mellitus, Type 1/pathology , Female , Hyperinsulinism/immunology , Hyperinsulinism/pathology , Lymph Nodes/immunology , Lymph Nodes/pathology , Lymphocyte Activation , Male , Membrane Glycoproteins/analysis , Membrane Glycoproteins/physiology , Mice , Mice, Inbred NOD/genetics , Mice, SCID , Mice, Transgenic , Pancreas/pathology , Peptides/analysis , Peptides/physiology , Plasmids , Polymerase Chain Reaction , Signal Transduction , T-Lymphocytes/immunology
14.
Exp Cell Res ; 314(3): 421-9, 2008 Feb 01.
Article in English | MEDLINE | ID: mdl-18164704

ABSTRACT

Angiogenesis, the process of new blood vessel formation, is important in wound healing, inflammation, tumorigenesis and metastases. During this process, it is a critical step of the loosening of cellular interactions between endothelial cells, which are dependent on the architecture of adherens junction constructed by homophilic interactions of cell surface cadherins. Several studies suggested that the dynamic changes of cadherins are necessary during angiogenesis. However, the mechanism of cadherins regulation on endothelial cells requires further delineation. Here, we showed that basic fibroblast growth factor (bFGF), a pivotal pro-angiogenic factor, can downregulate typical cadherins (E-, N-, P- and VE-cadherin) expression on the surface of human umbilical vein endothelial cells (HUVECs) via FGF receptor 1 (FGFR1) signaling. The bFGF-mediated surface cadherin downregulation was significantly reversed only when the HUVECs were treated with JNK inhibitor (SP600125), but not ERK (PD98059) or p38 inhibitor (SB203580). Infecting HUVECs with a dominant negative H-Ras mutant (Ras(S17N)) interferes bFGF-mediated cadherin downregulation, and the result suggests that bFGF attenuates surface cadherin expression on HUVECs via FGFR1 and intracellular Ras-JNK signaling. However, after growth factors withdrawal, FGFR1 blockade or JNK inhibition for 16 h, cadherins were re-expressed on cell surface of HUVECs. But the mRNA or total protein of cadherins had no significant change, suggesting that the effect of bFGF on cadherin expression may work through a post-translational control. Our data first suggest that JNK participates in bFGF-mediated surface cadherin downregulation. Loss of surface cadherins may affect the cell-cell interaction between endothelial cells and facilitate angiogenesis.


Subject(s)
Adherens Junctions/metabolism , Cadherins/metabolism , Endothelium, Vascular/metabolism , Fibroblast Growth Factor 2/metabolism , JNK Mitogen-Activated Protein Kinases/metabolism , Neovascularization, Physiologic/physiology , Adherens Junctions/drug effects , Adherens Junctions/ultrastructure , Anthracenes/pharmacology , Cadherins/genetics , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Communication/physiology , Cells, Cultured , Down-Regulation/physiology , Endothelium, Vascular/drug effects , Endothelium, Vascular/ultrastructure , Enzyme Inhibitors/pharmacology , Fibroblast Growth Factor 2/pharmacology , Humans , Infant, Newborn , JNK Mitogen-Activated Protein Kinases/antagonists & inhibitors , Protein Processing, Post-Translational/drug effects , Protein Processing, Post-Translational/physiology , RNA, Messenger/drug effects , RNA, Messenger/metabolism , Receptor, Fibroblast Growth Factor, Type 1/agonists , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Signal Transduction/physiology , ras Guanine Nucleotide Exchange Factors/genetics
15.
Ann N Y Acad Sci ; 1042: 325-37, 2005 May.
Article in English | MEDLINE | ID: mdl-15965078

ABSTRACT

We investigated whether H2O2, superoxide, and ERK participate in nerve growth factor (NGF)-induced signaling cascades and whether antioxidant N-acetylcysteine (NAC) regulates these NGF-induced responses. PC12 cells were cultured in medium containing NGF or vehicle with or without NAC pretreatment, and the intracellular H2O2 and superoxide levels and the amount of phosphorylated ERK were evaluated by flow cytometry and Western blotting, respectively. We found that NGF increased intracellular H2O2 concentration and activated ERK but failed to affect intracellular superoxide level. Moreover, NAC counteracted these NGF-induced responses. These findings demonstrate that NAC blocks the NGF-induced H2O2/ERK signaling in PC12 cells.


Subject(s)
Acetylcysteine/pharmacology , Antioxidants/pharmacology , Hydrogen Peroxide/metabolism , MAP Kinase Signaling System/drug effects , Nerve Growth Factor/pharmacology , Animals , Mitogen-Activated Protein Kinase 1/antagonists & inhibitors , Mitogen-Activated Protein Kinase 1/metabolism , Mitogen-Activated Protein Kinase 3/antagonists & inhibitors , Mitogen-Activated Protein Kinase 3/metabolism , PC12 Cells , Phosphorylation/drug effects , Protein Kinase Inhibitors/pharmacology , Rats , Superoxides/metabolism
16.
J Biomed Sci ; 10(6 Pt 2): 725-30, 2003.
Article in English | MEDLINE | ID: mdl-14631111

ABSTRACT

Adenosine triphosphate (ATP)-MgCl(2) attenuates ischemia-reperfusion (I-R)-induced lung injury in rats. A previous study indirectly suggests that Mg(2+)-dependent ecto-ATPases on the surface of leukocytes are responsible for the hydrolysis of ATP-MgCl(2) to adenosine, which then contributes to the protective effect of ATP-MgCl(2). This study investigated the role of leukocytes in I-R injury and the protective effect of ATP-MgCl(2) in our buffer-perfused isolated rat lung model. After isolating the lung blood flow of adult male Sprague-Dawley rats, the lungs were perfused through the pulmonary artery cannula with a physiologic salt solution containing human serum albumin. The protective effect of ATP-MgCl(2) pretreatment with or without leukocytes was investigated. Capillary permeability (K(fc)), lung weight gain (LWG), lung wet weight/body weight ratio (LW/BW), lung lavage protein concentration (LPC) and pulmonary artery pressure (PAP) were measured. I-R produced a significant increase in K(fc), LWG, LW/BW, LPC, and PAP. The increases in these indices were significantly attenuated by pretreatment with ATP-MgCl(2) (1 x 10(-6)M) together with leukocytes (2.9 x 10(6)/ml in the perfusate) but not with ATP-MgCl(2) alone. Our data suggest that I-R-induced acute lung injury is not dependent on circulating leukocytes. Pretreatment with ATP-MgCl(2) plus leukocytes but not ATP-MgCl(2) alone had protective effects against I-R lung injury. Whether these findings occur in vivo could not be determined in this study. In our isolated lung red blood cell-free perfusate system, the protective effect of ATP-MgCl(2) requires the presence of leukocytes.


Subject(s)
Adenosine Triphosphate/pharmacology , Adenosine Triphosphate/physiology , Leukocytes/physiology , Lung/drug effects , Reperfusion Injury/physiopathology , Animals , Bronchoalveolar Lavage Fluid/chemistry , Capillary Permeability/drug effects , Lung/physiopathology , Male , Perfusion , Pulmonary Wedge Pressure , Rats , Rats, Sprague-Dawley
SELECTION OF CITATIONS
SEARCH DETAIL