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










Publication year range
1.
Folia Histochem Cytobiol ; 56(4): 215-221, 2018.
Article in English | MEDLINE | ID: mdl-30565205

ABSTRACT

INTRODUCTION: Spinal muscular atrophy (SMA) is one of the most common genetic causes of death in infants due to a mutation of the motor neuron 1 (SMN1) gene. The SMN1 gene encodes for the multifunctional SMN protein. SMN has been shown to be implicated in pre-mRNA splicing, mRNA transport and translational control. Also other mRNA processing proteins, such as GLE1, Marten (MART3) and Fused in Sarcoma (FUS), have been linked to neurodegenerative diseases. The aim of the study was to determine the expression of SMN, GLE1, MART3 and FUS genes in cell lines of the fibroblasts derived from SMA patients and normal controls. MATERIAL AND METHODS: Total RNA was extracted from purchased fibroblasts acquired from three SMA type I patients and fibroblasts of three age-matched healthy controls. The RNA was then subjected to qPCR analysis using primers specific for the GLE1, MART3, FUS and SMN1 genes vs. GAPDH as internal control gene. RESULTS: SMN1 mRNA levels were at least ×10 lower in fibroblasts of SMA patients compared to controls. Gle1 and MART3 gene expression was ×2 downregulated whereas FUS mRNA levels appeared to be ×3 upregulated in SMA cells when compared to controls. We found a high correlation between FUS gene expression level to the SMN1 at gene expression level of fibroblast cell lines of SMA type I patients (r = 0.994, p < 0.0001). CONCLUSIONS: Our preliminary data show an intriguing expression profile of Gle1, MART3 and FUS genes in SMA, and suggest a critical role of FUS protein in the SMA pathogenesis.


Subject(s)
Fibroblasts/metabolism , Gene Expression Regulation , Muscular Atrophy, Spinal/metabolism , Nucleocytoplasmic Transport Proteins/biosynthesis , RNA-Binding Protein FUS/biosynthesis , Survival of Motor Neuron 1 Protein/biosynthesis , Cell Line , Fibroblasts/pathology , Humans , Muscular Atrophy, Spinal/pathology
2.
Anticancer Res ; 37(8): 4093-4101, 2017 08.
Article in English | MEDLINE | ID: mdl-28739693

ABSTRACT

BACKGROUND/AIM: We investigated the relationship between the expression of natural killer group 2, member D ligands (NKG2DLs) and the antitumor effects of protein-bound polysaccharide-K (PSK). MATERIALS AND METHODS: PSK was administered to evaluate its effectiveness against tumor growth. The expression of Rae-1 and H60 were analyzed in multiple cell lines. RESULTS: PSK showed the highest antitumor effects in mice implanted with cells expressing neither Rae-1 nor H60. PSK had little antitumor effect in mice implanted with cells expressing both Rae-1 and H60. A correlation between the expression of NKG2DLs and the antitumor effect of PSK was observed. After PSK administration, INF-γ production in CD8+ T cells increased in mice with cells expressing neither Rae-1 nor H60, but did not change in mice implanted with cells expressing both Rae-1 and H60. CONCLUSION: We demonstrated that the expression of NKG2DLs affects tumor immunity and the efficacy of immuno therapy in tumor-bearing mouse model.


Subject(s)
Fungal Proteins/administration & dosage , Minor Histocompatibility Antigens/genetics , NK Cell Lectin-Like Receptor Subfamily K/genetics , Neoplasms/drug therapy , Nuclear Matrix-Associated Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics , Polysaccharides/administration & dosage , Animals , CD8-Positive T-Lymphocytes/drug effects , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Ligands , Mice , Minor Histocompatibility Antigens/biosynthesis , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Neoplasms/genetics , Neoplasms/pathology , Nuclear Matrix-Associated Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/biosynthesis , Xenograft Model Antitumor Assays
3.
J Cancer Res Clin Oncol ; 142(11): 2309-17, 2016 Nov.
Article in English | MEDLINE | ID: mdl-27601163

ABSTRACT

PURPOSE: Aberrant expression of eukaryotic initiation factor 4E (eIF4E) has been observed in human malignancies. However, its role in hepatocellular carcinoma (HCC) remains to be established. The purpose of this study was to detect eIF4E expression and to evaluate its clinical relevance. METHODS: The eIF4E expression was studied in ninety HCC and randomly selected thirty-one non-tumor tissues from the same patient cohort, as well as in normal hepatic and HCC cell lines. The relation between its expression and clinicopathological parameters was also analyzed. RESULTS: eIF4E expression was higher in HCC samples and cell lines compared with that in non-tumor tissues (P < 0.001) and hepatocyte LO2, respectively. eIF4E overexpression was significantly associated with tumor number (P = 0.005) and incomplete encapsulation (P = 0.001). The 5-year overall survival rate and disease-free survival rate for patients with high eIF4E expression were 32.5 and 31.2 %, respectively; and for low eIF4E expression, it was 67.9 and 64.4 %, respectively (P < 0.001). Furthermore, subgroup analysis showed that high eIF4E level predicted poorer overall survival only for incomplete encapsulation (P = 0.001) and cirrhosis (P < 0.001), but not for complete encapsulation (P = 0.804) and non-cirrhosis (P = 0.359). Multivariate analysis revealed that eIF4E overexpression was an independent indicator for both overall survival (hazard ratio, 2.015; P = 0.043) and disease-free survival (hazard ratio, 2.666; P = 0.006). CONCLUSIONS: eIF4E protein might result in the malignant progression of HCC, and its overexpression may be a powerful prognostic biomarker and therapeutic target for HCC patients.


Subject(s)
Carcinoma, Hepatocellular/metabolism , Liver Neoplasms/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Carcinoma, Hepatocellular/pathology , Cell Line, Tumor , Disease Progression , Female , Humans , Immunohistochemistry , Kaplan-Meier Estimate , Liver Neoplasms/pathology , Male , Middle Aged , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Nucleocytoplasmic Transport Proteins/biosynthesis , Prognosis , Survival Rate , Tissue Array Analysis , Up-Regulation
4.
PLoS Genet ; 12(8): e1006198, 2016 08.
Article in English | MEDLINE | ID: mdl-27494403

ABSTRACT

Hippo signaling acts as a master regulatory pathway controlling growth, proliferation, and apoptosis and also ensures that variations in proliferation do not alter organ size. How the pathway coordinates restricting proliferation with organ size control remains a major unanswered question. Here we identify Rae1 as a highly-conserved target of the Hippo Pathway integrating proliferation and organ size. Genetic and biochemical studies in Drosophila cells and tissues and in mammalian cells indicate that Hippo signaling promotes Rae1 degradation downstream of Warts/Lats. In proliferating cells, Rae1 loss restricts cyclin B levels and organ size while Rae1 over-expression increases cyclin B levels and organ size, similar to Hippo Pathway over-activation or loss-of-function, respectively. Importantly, Rae1 regulation by the Hippo Pathway is crucial for its regulation of cyclin B and organ size; reducing Rae1 blocks cyclin B accumulation and suppresses overgrowth caused by Hippo Pathway loss. Surprisingly, in addition to suppressing overgrowth, reducing Rae1 also compromises survival of epithelial tissue overgrowing due to loss of Hippo signaling leading to a tissue "synthetic lethality" phenotype. Excitingly, Rae1 plays a highly conserved role to reduce the levels and activity of the Yki/YAP oncogene. Rae1 increases activation of the core kinases Hippo and Warts and plays a post-transcriptional role to increase the protein levels of the Merlin, Hippo, and Warts components of the pathway; therefore, in addition to Rae1 coordinating organ size regulation with proliferative control, we propose that Rae1 also acts in a feedback circuit to regulate pathway homeostasis.


Subject(s)
Drosophila Proteins/genetics , Intracellular Signaling Peptides and Proteins/genetics , Neurofibromin 2/genetics , Nuclear Matrix-Associated Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics , Protein Kinases/genetics , Protein Serine-Threonine Kinases/genetics , Animals , Apoptosis/genetics , Cell Proliferation/genetics , Cyclin B/genetics , Drosophila/genetics , Drosophila/growth & development , Drosophila Proteins/biosynthesis , Gene Expression Regulation, Developmental , Intracellular Signaling Peptides and Proteins/biosynthesis , Mitosis/genetics , Neurofibromin 2/biosynthesis , Nuclear Matrix-Associated Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/biosynthesis , Organ Size , Phenotype , Protein Kinases/biosynthesis , Protein Serine-Threonine Kinases/biosynthesis , Signal Transduction , Synthetic Lethal Mutations/genetics , Wings, Animal/growth & development
5.
Oncotarget ; 6(26): 22799-811, 2015 Sep 08.
Article in English | MEDLINE | ID: mdl-26078354

ABSTRACT

Protein arginine methyltransferases (PRMTs) plays critical roles in cancer. PRMT5 has been implicated in several types of tumors. However, the role of PRMT5 in cancer development remains to be fully elucidated. Here, we provide evidence that PRMT5 is overexpressed in colorectal cancer (CRC) cells and patient-derived primary tumors, correlated with increased cell growth and decreased overall patient survival. Arginine methyltransferase inhibitor 1 (AMI-1)strongly inhibited tumor growth, increased the ratio of Bax/Bcl-2, and induced apoptosis in mouse CRC xenograt model. AMI-1 also induced apoptosis and decreased the migratory activity in several CRC cells. In CRC xenografts AMI-1 significantly decreased symmetric dimethylation of histone 4 (H4R3me2s), a histone mark of type II PRMT5, but not the expression of H4R3me2a, a histone mark of type I PRMTs. These results suggest that the inhibition of PRMT5 contributes to the antitumor efficacy of AMI-1. Chromatin immunoprecipitation (ChIP) identified FGFR3 and eIF4E as two key genes regulated by PRMT5. PRMT5 knockdown reduced the levels of H4R3me2s and H3R8me2s methylation on FGFR3 and eIF4E promoters, leading to decreased expressions of FGFR3 and eIF4E. Collectively, our findings provide new evidence that PRMT5 plays an important role in CRC pathogenesis through epigenetically regulating arginine methylation of oncogenes such as eIF4E and FGFR3.


Subject(s)
Colorectal Neoplasms/metabolism , Colorectal Neoplasms/therapy , Naphthalenesulfonates/pharmacology , Nucleocytoplasmic Transport Proteins/metabolism , Protein-Arginine N-Methyltransferases/metabolism , Receptor, Fibroblast Growth Factor, Type 3/metabolism , Urea/analogs & derivatives , Animals , Arginine/genetics , Arginine/metabolism , Cell Proliferation/physiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/pathology , Female , Gene Knockdown Techniques , HCT116 Cells , Humans , Male , Methylation , Mice , Mice, Inbred BALB C , Mice, Nude , Middle Aged , Molecular Targeted Therapy , Nucleocytoplasmic Transport Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/genetics , Protein-Arginine N-Methyltransferases/antagonists & inhibitors , Protein-Arginine N-Methyltransferases/genetics , RNA, Small Interfering/administration & dosage , RNA, Small Interfering/genetics , Receptor, Fibroblast Growth Factor, Type 3/genetics , Transfection , Urea/pharmacology , Xenograft Model Antitumor Assays
6.
J Immunol ; 194(11): 5539-48, 2015 Jun 01.
Article in English | MEDLINE | ID: mdl-25911757

ABSTRACT

S100A8/A9, a proinflammatory protein, is upregulated in inflammatory diseases, and also has a tumor-promoting activity by the recruitment of myeloid cells and tumor cell invasion. However, whether the expression of S100A8/A9 in tumors predicts a good or poor prognosis is controversial in the clinical setting. In this study, to clarify the in vivo role of S100A8/A9 in the tumor microenvironment, we s.c. inoculated Pan02 cells stably expressing S100A8 and S100A9 proteins (Pan02-S100A8/A9) in syngeneic C57BL/6 mice. Unexpectedly, after small tumor nodules were once established, they rapidly disappeared. Flow cytometry showed that the number of NK cells in the tumors was increased, and an administration of anti-asialoGM1 Ab for NK cell depletion promoted the growth of Pan02-S100A8/A9 s.c. tumors. Although the S100A8/A9 proteins alone did not change the IFN-γ expression of NK cells in vitro, a coculture with Pan02 cells, which express Rae-1, induced IFN-γ production, and Pan02-S100A8/A9 cells further increased the number of IFN-γ(+) NK cells, suggesting that S100A8/A9 enhanced the NK group 2D ligand-mediated intracellular activation pathway in NK cells. We then examined whether NK cell activation by S100A8/A9 was via their binding to receptor of advanced glycation end product (RAGE) by using the inhibitors. RAGE antagonistic peptide and anti-RAGE Ab inhibited the IFN-γ production of NK cells induced by S100A8/A9 proteins, and an administration of FPS-ZM1, a RAGE inhibitor, significantly enhanced the in vivo growth of Pan02-S100A8/A9 tumors. We thus found a novel activation mechanism of NK cells via S100A8/A9-RAGE signaling, which may open a novel perspective on the in vivo interaction between inflammation and innate immunity.


Subject(s)
Calgranulin A/immunology , Calgranulin B/immunology , Interferon-gamma/biosynthesis , Killer Cells, Natural/immunology , Pancreatic Neoplasms/immunology , Receptors, Immunologic/immunology , Animals , Benzamides/pharmacology , Calgranulin A/biosynthesis , Calgranulin B/biosynthesis , Cell Line, Tumor , Cell Proliferation , Female , Inflammation/immunology , Interferon-gamma/immunology , Lymphocyte Activation/immunology , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Nude , Neoplasm Transplantation , Nuclear Matrix-Associated Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/biosynthesis , Receptor for Advanced Glycation End Products , Receptors, Immunologic/antagonists & inhibitors , Transplantation, Isogeneic , Tumor Microenvironment/immunology
7.
Asian Pac J Cancer Prev ; 15(22): 9667-72, 2014.
Article in English | MEDLINE | ID: mdl-25520086

ABSTRACT

BACKGROUND: Pancreatic adenocarcinoma is a malignant gastrointestinal cancer with significant morbidity and mortality. Despite severe side effects of chemotherapy, the use of immunotherapy combined with chemotherapy has emerged as a common clinical treatment. In this study, we investigated the efficacy of the combined doxorubicin and interferon-α (IFN-α) therapy on murine pancreatic cancer Panc02 cells in vitro and in vivo and underlying mechanisms. MATERIALS AND METHODS: A Panc02-bearing mouse model was established to determine whether doxorubicin and interferon-α (IFN-α) could effectively inhibit tumor growth in vivo. Cytotoxicity of natural killer (NK) cells and cytotoxic T lymphocytes (CTLs) was evaluated using a standard LDH release assay. To evaluate the relevance of NK cells and CD8 T cells to the combination therapy-mediated anti-tumor effects, they were depleted in tumor-bearing mice by injecting anti-asialo-GM-1 antibodies or anti-CD8 antibodies, respectively. Finally, the influence of doxorubicin+interferon-α (IFN-α) on the ligands of NK and T cells was assessed by flow cytometry. RESULTS: The combination therapy group demonstrated a significant inhibition of growth of Panc02 in vivo, resulting from activated cytotoxicity of NK cells and CTLs. Depleting CD8 T cells or NK cells reduced the anticancer effects mediated by immunochemotherapy. Furthermore, the doxorubicin+IFN-a treatment increased the expression of major histocompatibility complex class I (MHC I) and NKG2D ligands on Panc02 cells, suggesting that the combined therapy may be a potential strategy for enhancing immunogenicity of tumors. All these data indicate that the combination therapy using doxorubicin and interferon-α (IFN-α) may be a potential strategy for treating pancreatic adenocarcinoma.


Subject(s)
Doxorubicin/pharmacology , Histocompatibility Antigens Class I/biosynthesis , Interferon-alpha/pharmacology , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Pancreatic Neoplasms/drug therapy , Animals , Antibiotics, Antineoplastic/pharmacology , Antineoplastic Agents/pharmacology , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Carrier Proteins/biosynthesis , Cell Line, Tumor , Immunotherapy/methods , Killer Cells, Natural/immunology , Lymphocyte Depletion , Male , Membrane Proteins , Mice , Mice, Inbred C57BL , Neoplasm Transplantation , Nuclear Matrix-Associated Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/biosynthesis , Pancreatic Neoplasms/immunology , T-Lymphocytes, Cytotoxic/immunology , Up-Regulation/drug effects
8.
Arch Biochem Biophys ; 564: 128-35, 2014 Dec 15.
Article in English | MEDLINE | ID: mdl-25303791

ABSTRACT

Tripeptidyl-peptidase II (TPPII) is an aminopeptidase with suggested regulatory effects on cell cycle, apoptosis and senescence. A protein-protein interaction study revealed that TPPII physically interacts with the tumor suppressor MYBBP1A and the cell cycle regulator protein CDK2. Mutual protein-protein interaction was detected between MYBBP1A and CDK2 as well. In situ Proximity Ligation Assay (PLA) using HEK293 cells overexpressing TPPII forming highly enzymatically active oligomeric complexes showed that the cytoplasmic interaction frequency of TPPII with MYBBP1A increased with the protein expression of TPPII and using serum-free cell growth conditions. A specific reversible inhibitor of TPPII, butabindide, suppressed the cytoplasmic interactions of TPPII and MYBBP1A both in control HEK293 and the cells overexpressing murine TPPII. The interaction of MYBBP1A with CDK2 was confirmed by in situ PLA in two different mammalian cell lines. Functional link between TPPII and MYBBP1A has been verified by gene expression study during anoikis, where overexpression of TPP II decreased mRNA expression level of MYBBP1A at the cell detachment conditions. All three interacting proteins TPPII, MYBBP1A and CDK2 have been previously implicated in the research for development of tumor-suppressing agents. This is the first report presenting mutual protein-protein interaction network of these proteins.


Subject(s)
Aminopeptidases/biosynthesis , Cyclin-Dependent Kinase 2/metabolism , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/biosynthesis , Gene Expression Regulation/physiology , Nuclear Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/biosynthesis , Protein Biosynthesis/physiology , Serine Endopeptidases/biosynthesis , Aminopeptidases/antagonists & inhibitors , Aminopeptidases/genetics , Anoikis/drug effects , Anoikis/physiology , Cyclin-Dependent Kinase 2/genetics , DNA-Binding Proteins , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/antagonists & inhibitors , Dipeptidyl-Peptidases and Tripeptidyl-Peptidases/genetics , Gene Expression Regulation/drug effects , HEK293 Cells , Humans , Indoles/pharmacology , K562 Cells , Nuclear Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics , Protein Biosynthesis/drug effects , RNA-Binding Proteins , Serine Endopeptidases/genetics , Transcription Factors
9.
Cancer Res ; 74(8): 2193-2203, 2014 Apr 15.
Article in English | MEDLINE | ID: mdl-24590060

ABSTRACT

The immunoreceptor NKG2D originally identified in natural killer (NK) cells recognizes ligands that are upregulated on tumor cells. Expression of NKG2D ligands (NKG2DL) is induced by the DNA damage response (DDR), which is often activated constitutively in cancer cells, revealing them to NK cells as a mechanism of immunosurveillance. Here, we report that the induction of retinoic acid early transcript 1 (RAE1) ligands for NKG2D by the DDR relies on a STING-dependent DNA sensor pathway involving the effector molecules TBK1 and IRF3. Cytosolic DNA was detected in lymphoma cell lines that express RAE1 and its occurrence required activation of the DDR. Transfection of DNA into ligand-negative cells was sufficient to induce RAE1 expression. Irf3(+/-);Eµ-Myc mice expressed lower levels of RAE1 on tumor cells and showed a reduced survival rate compared with Irf3(+/+);Eµ-Myc mice. Taken together, our results suggest that genomic damage in tumor cells leads to activation of STING-dependent DNA sensor pathways, thereby activating RAE1 and enabling tumor immunosurveillance.


Subject(s)
DNA Damage/genetics , DNA Damage/physiology , DNA, Neoplasm/metabolism , Lymphoma/metabolism , Membrane Proteins/metabolism , NK Cell Lectin-Like Receptor Subfamily K/metabolism , Nuclear Matrix-Associated Proteins/metabolism , Nucleocytoplasmic Transport Proteins/metabolism , Animals , Cell Line, Tumor , DNA Damage/radiation effects , Immunologic Surveillance , Interferon Regulatory Factor-3/genetics , Interferon Regulatory Factor-3/immunology , Interferon Regulatory Factor-3/metabolism , Killer Cells, Natural/immunology , Killer Cells, Natural/metabolism , Ligands , Lymphoma/genetics , Lymphoma/immunology , Membrane Proteins/genetics , Membrane Proteins/immunology , Mice , Mice, Inbred C57BL , Mice, Transgenic , NK Cell Lectin-Like Receptor Subfamily K/genetics , NK Cell Lectin-Like Receptor Subfamily K/immunology , Nuclear Matrix-Associated Proteins/biosynthesis , Nuclear Matrix-Associated Proteins/genetics , Nuclear Matrix-Associated Proteins/immunology , Nucleocytoplasmic Transport Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/genetics , Nucleocytoplasmic Transport Proteins/immunology , Phosphorylation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/immunology , Protein Serine-Threonine Kinases/metabolism , Transfection , Up-Regulation
10.
Mol Cancer ; 12(1): 166, 2013 Dec 18.
Article in English | MEDLINE | ID: mdl-24350772

ABSTRACT

BACKGROUND: Cancer is both a systemic and a genetic disease. The pathogenesis of cancer might be related to dampened immunity. Host immunity recognizes nascent malignant cells - a process referred to as immune surveillance. Augmenting immune surveillance and suppressing immune escape are crucial in tumor immunotherapy. METHODS: A recombinant plasmid capable of co-expressing granulocyte-macrophage colony- stimulating factor (GM-SCF), interleukin-21 (IL-21), and retinoic acid early transcription factor-1 (Rae-1) was constructed, and its effects determined in a mouse model of subcutaneous liver cancer. Serum specimens were assayed for IL-2 and INF-γ by ELISA. Liver cancer specimens were isolated for Rae-1 expression by RT-PCR and Western blot, and splenocytes were analyzed by flow cytometry. RESULTS: The recombinant plasmid inhibited the growth of liver cancer and prolonged survival of tumor-loaded mice. Activation of host immunity might have contributed to this effect by promoting increased numbers and cytotoxicity of natural killer (NK) cells and cytotoxic T lymphocytes (CTL) following expression of GM-SCF, IL-21, and Rae-1. By contrast, the frequency of regulatory T cells was decreased, Consequently, activated CTL and NK cells enhanced their secretion of INF-γ, which promoted cytotoxicity of NK cells and CTL. Moreover, active CTL showed dramatic secretion of IL-2, which stimulates CTL. The recombinant expression plasmid also augmented Rae-1 expression by liver cancer cells. Rae-1 receptor expressing CTL and NK cells removed liver cancer. CONCLUSIONS: The recombinant expression plasmid inhibited liver cancer by a mechanism that involved activation of cell-mediated immunity and Rae-1 in liver cancer.


Subject(s)
Genetic Therapy , Granulocyte-Macrophage Colony-Stimulating Factor/genetics , Immunity, Cellular , Interleukins/genetics , Liver Neoplasms, Experimental/immunology , Nuclear Matrix-Associated Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics , Animals , Cell Line, Tumor , Granulocyte-Macrophage Colony-Stimulating Factor/biosynthesis , Humans , Interferon-gamma/blood , Interleukin-2/blood , Interleukins/biosynthesis , Killer Cells, Natural/immunology , Liver Neoplasms, Experimental/metabolism , Liver Neoplasms, Experimental/therapy , Male , Mice , Mice, Inbred BALB C , Neoplasm Transplantation , Nuclear Matrix-Associated Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/biosynthesis , T-Lymphocytes, Cytotoxic/immunology
11.
J Autoimmun ; 40: 66-73, 2013 Feb.
Article in English | MEDLINE | ID: mdl-22944096

ABSTRACT

It is thought that viral infections might jeopardize regulatory T cell therapy in type 1 diabetes. Viral infections can lead to surface expression of ligands for the activating NKG2D receptor, such as retinoic acid early transcript 1 (Rae-1), whose expression on beta-cells recruits NKG2D(+) autoreactive CD8(+) T cells. Both in men and mice, autoreactive cytotoxic T cells express NKG2D. We showed that NKG2D expression increased on CD4(+) and CD8(+) T cells during virus-induced diabetes development in the rat insulin promotor (RIP) Lymphocytic Choriomeningitis Virus (LCMV) model. Combination treatment with anti-NKG2D and antigen-specific regulatory T cells (Treg), at doses inefficacious in mono-treatment, synergized to prevent diabetes in 75% of the virus-infected RIP-LCMV mice. Nevertheless, NKG2D blockade alone failed to reverse recent-onset diabetes in non-obese diabetic (NOD) mice, despite downregulation of NKG2D on NK cells in the blood and CD8(+) T cells in the spleen and pancreatic lymph nodes. Our data suggest that blocking the interaction of NKG2D with it ligands is insufficient to protect against diabetes when a strong inflammatory process actively drives NKG2D upregulation, but should be considered to help maintaining Treg functionality during ongoing pancreatic inflammation.


Subject(s)
Diabetes Mellitus, Type 1/prevention & control , Lymphocytic Choriomeningitis/immunology , Lymphocytic choriomeningitis virus/immunology , NK Cell Lectin-Like Receptor Subfamily K/metabolism , T-Lymphocytes, Regulatory/immunology , Animals , Antibodies/immunology , CD8-Positive T-Lymphocytes/immunology , Diabetes Mellitus, Type 1/immunology , Diabetes Mellitus, Type 1/virology , Killer Cells, Natural/immunology , Lymph Nodes/immunology , Mice , Mice, Inbred NOD , NK Cell Lectin-Like Receptor Subfamily K/immunology , Nuclear Matrix-Associated Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/biosynthesis , Pancreas/immunology , Pancreas/virology , Spleen/immunology
12.
J Clin Invest ; 122(10): 3718-30, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22945631

ABSTRACT

A promising strategy for cancer immunotherapy is to disrupt key pathways regulating immune tolerance, such as cytotoxic T lymphocyte-associated protein 4 (CTLA-4). However, the determinants of response to anti-CTLA-4 mAb treatment remain incompletely understood. In murine models, anti-CTLA-4 mAbs alone fail to induce effective immune responses to poorly immunogenic tumors but are successful when combined with additional interventions, including local ionizing radiation (IR) therapy. We employed an established model based on control of a mouse carcinoma cell line to study endogenous tumor-infiltrating CD8+ T lymphocytes (TILs) following treatment with the anti-CTLA-4 mAb 9H10. Alone, 9H10 monotherapy reversed the arrest of TILs with carcinoma cells in vivo. In contrast, the combination of 9H10 and IR restored MHC class I-dependent arrest. After implantation, the carcinoma cells had reduced expression of retinoic acid early inducible-1 (RAE-1), a ligand for natural killer cell group 2D (NKG2D) receptor. We found that RAE-1 expression was induced by IR in vivo and that anti-NKG2D mAb blocked the TIL arrest induced by IR/9H10 combination therapy. These results demonstrate that anti-CTLA-4 mAb therapy induces motility of TIL and that NKG2D ligation offsets this effect to enhance TILs arrest and antitumor activity.


Subject(s)
Antibodies, Monoclonal/therapeutic use , CD8-Positive T-Lymphocytes/drug effects , CTLA-4 Antigen/antagonists & inhibitors , Immunotherapy , Lymphocytes, Tumor-Infiltrating/drug effects , Mammary Neoplasms, Experimental/therapy , Animals , Antibodies, Monoclonal/pharmacology , CD8-Positive T-Lymphocytes/immunology , CD8-Positive T-Lymphocytes/radiation effects , Cell Line, Tumor/immunology , Cell Line, Tumor/transplantation , Cell Movement , Combined Modality Therapy , Cytotoxicity, Immunologic/drug effects , Cytotoxicity, Immunologic/radiation effects , Drug Screening Assays, Antitumor , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/radiation effects , H-2 Antigens/immunology , Intercellular Adhesion Molecule-1/biosynthesis , Intercellular Adhesion Molecule-1/genetics , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/radiation effects , Mammary Neoplasms, Experimental/immunology , Mammary Neoplasms, Experimental/radiotherapy , Mammary Neoplasms, Experimental/secondary , Mice , Mice, Inbred BALB C , NK Cell Lectin-Like Receptor Subfamily K/antagonists & inhibitors , NK Cell Lectin-Like Receptor Subfamily K/immunology , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , Nuclear Matrix-Associated Proteins/biosynthesis , Nuclear Matrix-Associated Proteins/genetics , Nucleocytoplasmic Transport Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/genetics , Receptors, CXCR/genetics , Receptors, CXCR6 , Tumor Microenvironment/immunology
13.
PLoS Pathog ; 7(9): e1002265, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21966273

ABSTRACT

Natural killer (NK) cells are lymphocytes that play a major role in the elimination of virally-infected cells and tumor cells. NK cells recognize and target abnormal cells through activation of stimulatory receptors such as NKG2D. NKG2D ligands are self-proteins, which are absent or expressed at low levels on healthy cells but are induced upon cellular stress, transformation, or viral infection. The exact molecular mechanisms driving expression of these ligands remain poorly understood. Here we show that murine cytomegalovirus (MCMV) infection activates the phosphatidylinositol-3-kinase (PI3K) pathway and that this activation is required for the induction of the RAE-1 family of mouse NKG2D ligands. Among the multiple PI3K catalytic subunits, inhibition of the p110α catalytic subunit blocks this induction. Similarly, inhibition of p110α PI3K reduces cell surface expression of RAE-1 on transformed cells. Many viruses manipulate the PI3K pathway, and tumors frequently mutate the p110α oncogene. Thus, our findings suggest that dysregulation of the PI3K pathway is an important signal to induce expression of RAE-1, and this may represent a commonality among various types of cellular stresses that result in the induction of NKG2D ligands.


Subject(s)
Cytomegalovirus Infections/physiopathology , Killer Cells, Natural/metabolism , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Nuclear Matrix-Associated Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/biosynthesis , Phosphatidylinositol 3-Kinase/physiology , Receptors, Natural Killer Cell/physiology , Animals , Catalytic Domain/physiology , Cell Line, Tumor , Cell Transformation, Viral , Class I Phosphatidylinositol 3-Kinases , Fibroblasts/virology , Killer Cells, Natural/immunology , Ligands , Mice , Muromegalovirus/immunology , Phosphatidylinositol 3-Kinases/physiology
14.
Nephrol Dial Transplant ; 26(12): 3873-81, 2011 Dec.
Article in English | MEDLINE | ID: mdl-21555390

ABSTRACT

BACKGROUND: Engagement of Toll-like receptor (TLR) 4 on intrinsic kidney cells is critical for the full development of renal ischaemia-reperfusion injury (IRI). Effects of TLR signalling in renal parenchymal cells include the production of cytokines, chemokines and other soluble mediators which contribute to local inflammation and leucocyte accumulation. Whether engagement of TLR4 on kidney cells results in additional pro-inflammatory modifications of the renal microenvironment remains to be determined. METHODS: Renal IRI was induced by clamping of the renal pedicles, and expression of NKG2D ligands in mice deficient in TLR4 or its adaptor molecule MyD88, or else pretreated with blocking antibodies against the endogenous TLR4 ligand HMGB1, was compared to that in wild-type mice. Cultures of isolated renal tubular epithelial cells (TECs) from WT, TLR4(-/-) and MyD88(-/-) mice were stimulated with the TLR4 ligand lipopolysaccharide (LPS), or mineral oil occlusion was used to simulate IRI in vitro, prior to determination of NKG2D ligand expression. Chimeric mice lacking TLR4 in either the bone marrow derived or the parenchymal compartment were also subjected to IRI. RESULTS: In this study, we demonstrate a substantial increase in the expression of the NKG2D ligands retinoic acid early inducible-1 (RAE-1), murine ULBP-like transcript 1 (MULT-1) and histocompatibility-60 (H-60) in mouse kidneys during renal IRI. Expression of NKG2D ligands was attenuated in mice deficient in either TLR4 or the adaptor molecule MyD88. Antibody blockade of HMGB1 reduced NKG2D ligand expression by a comparable extent to TLR4 deficiency and did not result in further reduction of NKG2D ligand expression in TLR4(-/-) mice. Isolated TECs from normal mice but not those with defects in the TLR4-MyD88 signalling pathway expressed RAE-1 and MULT-1 upon exposure to LPS and after being subjected to in vitro conditions resembling ischaemia-reperfusion. TLR4 competence in the parenchymal but not the bone marrow-derived compartment was required for RAE-1 up-regulation in mouse kidneys after ischaemia, while TLR4 signalling in both compartments contributed to the intrarenal expression of MULT-1 during IRI. CONCLUSION: Expression of the NKG2D ligands RAE-1 and MULT-1 on kidney cells in response to TLR4 engagement by HMGB1 represents another mechanism by which TLR4 signalling may participate in the pathogenesis of renal IRI.


Subject(s)
Epithelial Cells/metabolism , Kidney Tubules/cytology , NK Cell Lectin-Like Receptor Subfamily K/biosynthesis , Nuclear Matrix-Associated Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/biosynthesis , Toll-Like Receptor 4/physiology , Urothelium/cytology , Urothelium/metabolism , Animals , Ligands , Male , Mice
15.
J Biotechnol ; 153(3-4): 86-91, 2011 May 20.
Article in English | MEDLINE | ID: mdl-21473891

ABSTRACT

Recent research into mRNA maturation processes in the nucleus has identified a number of proteins involved in mRNA transcription, capping, splicing, end processing and export. Among them, the Tap-p15 heterodimer acts as an mRNA export receptor. Tap-p15 is recruited onto fully processed mRNA in the nucleus, which is ready for export to the cytoplasm, through associating with Aly or SR proteins on mRNA, or by directly associating with a constitutive transport element (CTE), an RNA element derived from type D retroviruses. mRNA containing a CTE is exported to the cytoplasm by directly associating with Tap-p15, even in the absence of Tap-recruiting proteins such as Aly or SR proteins on the mRNA. Here, we showed that the use of a CTE enhanced the expression of recombinant protein in human cell lines. The co-expression of reporter proteins and Tap-p15 also enhanced recombinant protein expression. Moreover, the use of a CTE and Tap-p15 synergistically further enhanced the recombinant protein expression. In addition to Tap-p15, several Tap-p15-recruiting proteins, including Aly and SR proteins, enhanced recombinant protein expression, albeit independently of the CTE. The incorporation of a CTE and Tap-p15-recruiting proteins into protein expression system is useful to increase recombinant protein yield in human cells.


Subject(s)
Nucleocytoplasmic Transport Proteins/biosynthesis , Protein Engineering/methods , RNA Transport , Recombinant Fusion Proteins/biosynthesis , Regulatory Sequences, Ribonucleic Acid , Alkaline Phosphatase/biosynthesis , Alkaline Phosphatase/chemistry , Alkaline Phosphatase/genetics , Blotting, Western , Cell Line, Tumor , Erythropoietin/biosynthesis , Erythropoietin/chemistry , Erythropoietin/genetics , Humans , Luciferases/biosynthesis , Luciferases/chemistry , Luciferases/genetics , Nucleocytoplasmic Transport Proteins/chemistry , Nucleocytoplasmic Transport Proteins/genetics , Recombinant Fusion Proteins/chemistry , Recombinant Fusion Proteins/genetics
16.
Exp Eye Res ; 89(6): 854-62, 2009 Dec.
Article in English | MEDLINE | ID: mdl-19627987

ABSTRACT

Previously we observed that avian corneal epithelial cells protect their DNA from oxidative damage by having the iron-sequestering molecule ferritin - normally cytoplasmic - in a nuclear location. This localization involves a developmentally-regulated ferritin-like protein - ferritoid - that initially serves as the nuclear transporter, and then as a component of a ferritoid-ferritin complex that is half the size of a typical ferritin and binds to DNA. We also observed that developmentally, the synthesis of ferritin and ferritoid are regulated coordinately - with ferritin being predominantly translational and ferritoid transcriptional. In the present study we examined whether the mechanism(s) involved in this regulation reside within the cornea itself, or alternatively involve a systemic factor(s). For this, we explanted embryonic corneas of one age to the chorioallantoic membrane (CAM) of host embryos of a different age - all prior to the initiation of ferritin synthesis. Consistent with systemic regulation, the explants initiated the synthesis of both ferritin and ferritoid in concert with that of the host. We then examined whether this systemic regulation might involve thyroxine - a hormone with broad developmental effects. Employing corneal organ cultures, we observed that thyroxine initiated the synthesis of both components in a manner similar to that which occurs in vivo (i.e. ferritin was translational and ferritoid transcriptional).


Subject(s)
DNA-Binding Proteins/biosynthesis , Epithelium, Corneal/metabolism , Eye Proteins/biosynthesis , Ferritins/biosynthesis , Nucleocytoplasmic Transport Proteins/biosynthesis , Thyroxine/physiology , Animals , Cell Nucleus/metabolism , Chick Embryo , Corneal Transplantation/methods , Culture Media, Serum-Free , Embryonic Development/physiology , Epithelium, Corneal/drug effects , Epithelium, Corneal/embryology , Organ Culture Techniques , Serum , Triiodothyronine/pharmacology
17.
Cancer Res ; 67(10): 5003-8, 2007 May 15.
Article in English | MEDLINE | ID: mdl-17510432

ABSTRACT

Despite advancements in the treatment of ovarian cancer, this disease continues to be a leading cause of cancer death in women. Adoptive transfer of tumor-reactive T cells is a promising antitumor therapy for many cancers. We designed a chimeric receptor linking NKG2D, a natural killer (NK) cell-activating receptor, to the CD3zeta chain of the T-cell receptor to target ovarian tumor cells. Engagement of chimeric NKG2D receptors (chNKG2D) with ligands for NKG2D, which are commonly expressed on tumor cells, leads to T-cell secretion of proinflammatory cytokines and tumor cytotoxicity. In this study, we show that >80% of primary human ovarian cancer samples expressed ligands for NKG2D on the cell surface. The tumor samples expressed MHC class I-related protein A, MICB, and UL-16 binding proteins 1 and 3. ChNKG2D-expressing T cells lysed ovarian cancer cell lines. We show that T cells from ovarian cancer patients that express chNKG2D secreted proinflammatory cytokines when cultured with autologous tumor cells. In addition, we show that chNKG2D T cells can be used therapeutically in a murine model of ovarian cancer. These data indicate that treatment with chNKG2D-expressing T cells is a potential immunotherapy for ovarian cancer.


Subject(s)
Immunotherapy, Adoptive/methods , Ovarian Neoplasms/immunology , Ovarian Neoplasms/therapy , Receptors, Immunologic/immunology , T-Lymphocytes/immunology , Animals , Cell Growth Processes/immunology , Cell Line, Tumor , Cytokines/immunology , Cytokines/metabolism , Female , Humans , Interferon-gamma/immunology , Interferon-gamma/metabolism , Mice , Mice, Inbred C57BL , NK Cell Lectin-Like Receptor Subfamily K , Nuclear Matrix-Associated Proteins/biosynthesis , Nuclear Matrix-Associated Proteins/immunology , Nucleocytoplasmic Transport Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/immunology , Receptors, Natural Killer Cell , T-Lymphocytes/metabolism
18.
Zhongguo Shi Yan Xue Ye Xue Za Zhi ; 15(1): 160-4, 2007 Feb.
Article in Chinese | MEDLINE | ID: mdl-17490545

ABSTRACT

The study was purposed to explore the effects of NKG2D receptor and its ligands RAE-1 and H60 on graft-versus-tumor (GVT) response induced by MHC haploidentical bone marrow/spleen cell transplantation. Female (BALB/c x C57BL/6) F1 mice (CB6F1, H-2K(b/d)) inoculated with H22 cells to develop a solid tumor model were the recipients, and bone marrow mixed with spleen cells of the healthy male C57BL/6 (H-2K(b)) mice were the donor cells. GVT response was observed after transplantation that from donor cells T and NK cells were purged with anti-CD3 and anti-NK monoclonal antibody, and the NKG2D receptor was blocked with anti-NKG2D monoclonal antibody, the expression levels of RAE-1 and H60 mRNA in tumor tissue were measured by means of semi-quantitative reverse transcription polymerase chain reaction (RT-PCR) at different time points after transplantation. The results showed that the GVT response of transplantation was reduced after in vitro depletion of T and NK cells or blocking NKG2D receptor in donor cells of the graft, the expression levels of RAE-1 and H60 mRNA in tumor tissue increased after transplantation of haploidential bone marrow mixed with spleen cells. It is concluded that NKG2D and its ligands RAE-1 and H60 may play important roles in GVT response.


Subject(s)
Graft vs Leukemia Effect/immunology , Hematopoietic Stem Cell Transplantation , Minor Histocompatibility Antigens/biosynthesis , Nuclear Matrix-Associated Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/biosynthesis , Receptors, Immunologic/blood , Animals , Female , Graft vs Leukemia Effect/drug effects , Killer Cells, Natural/drug effects , Killer Cells, Natural/immunology , Leukemia, Experimental/immunology , Leukemia, Experimental/therapy , Ligands , Male , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Mice, Inbred Strains , Minor Histocompatibility Antigens/genetics , NK Cell Lectin-Like Receptor Subfamily K , Nuclear Matrix-Associated Proteins/genetics , Nucleocytoplasmic Transport Proteins/genetics , Receptors, Immunologic/genetics , Receptors, Natural Killer Cell
19.
J Immunol ; 175(8): 5541-50, 2005 Oct 15.
Article in English | MEDLINE | ID: mdl-16210663

ABSTRACT

In an orthotopic murine model of head and neck squamous cell carcinoma (SCC VII/SF) we studied NK cell-mediated immunity following vaccination with a recombinant vaccinia virus expressing IL-2 (rvv-IL-2). SCC VII/SF tumor cells were injected into the oral cavity of C3H/HeJ mice on day 0. Mice were vaccinated on days 7, 10, and 14 with rvv-IL-2 and control vaccines. Phenotypes, numbers, and biological activities of NK cells were determined following vaccination. Levels of expression of NK-activating receptor NKG2D and CD16 on NK cell surface were assayed in the vaccinated mice. Expression of NKG2D ligands, Rae1, and H60 on SCC VII/SF cells was also examined. Vaccination with rvv-IL-2 resulted in expansion of NK cells. NK cells isolated from rvv-IL-2-vaccinated mice had significantly higher biological activities compared with mice treated with control vaccines. NK cells from tumor-bearing mice expressed significantly lower levels of NKG2D and CD16 compared with rvv-IL-2 vaccinated mice. SCC VII/SF tumors expressed NKG2D ligand Rae 1, although H60 was not present. SCC VII/SF tumors expressed high levels of TGF-beta1, which were down-modulated by vaccination with rvv-IL-2. Incubation of NK cells with tumor homogenate or cultured supernatant of SCC VII/SF cells reduced the expression of NKG2D and CD16. This inhibition appeared to be mediated by TGF-beta1. SCC VII/SF tumors in the oral cavity of the mice secrete high quantities of TGF-beta1, which reduce the expression of NK cell receptor NKG2D as well as CD16 and inhibits biological functions of NK cells.


Subject(s)
Head and Neck Neoplasms/immunology , Killer Cells, Natural/immunology , Receptors, Immunologic/antagonists & inhibitors , Transforming Growth Factor beta/physiology , Animals , Cells, Cultured , Cytotoxicity, Immunologic , Female , Head and Neck Neoplasms/metabolism , Immunization, Secondary , Killer Cells, Natural/metabolism , Ligands , Lymphocyte Count , Mice , Mice, Inbred C3H , Minor Histocompatibility Antigens/biosynthesis , Minor Histocompatibility Antigens/genetics , NK Cell Lectin-Like Receptor Subfamily K , Nuclear Matrix-Associated Proteins/biosynthesis , Nuclear Matrix-Associated Proteins/genetics , Nucleocytoplasmic Transport Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/genetics , Receptors, IgG/biosynthesis , Receptors, IgG/genetics , Receptors, Immunologic/genetics , Receptors, Immunologic/metabolism , Receptors, Natural Killer Cell , Reverse Transcriptase Polymerase Chain Reaction , Transforming Growth Factor beta1 , Tumor Cells, Cultured , Vaccinia virus/immunology , Viral Vaccines/immunology
20.
J Biol Chem ; 276(42): 38820-9, 2001 Oct 19.
Article in English | MEDLINE | ID: mdl-11489893

ABSTRACT

Nuclear transport factor 2 (NTF2) is a small homodimeric protein that interacts simultaneously with both RanGDP and FxFG nucleoporins. The interaction between NTF2 and Ran is essential for the import of Ran into the nucleus. Here we use mutational analysis to dissect the in vivo role of the interaction between NTF2 and nucleoporins. We identify a series of surface residues that form a hydrophobic patch on NTF2, which when mutated disrupt the NTF2-nucleoporin interaction. Analysis of these mutants in vivo demonstrates that the strength of this interaction can be significantly reduced without affecting cell viability. However, cells cease to be viable if the interaction between NTF2 and nucleoporins is abolished completely, indicating that this interaction is essential for the function of NTF2 in vivo. In addition, we have isolated a dominant negative mutant of NTF2, N77Y, which has increased affinity for nucleoporins. Overexpression of the N77Y protein blocks nuclear protein import and concentrates Ran at the nuclear rim. These data support a mechanism in which NTF2 interacts transiently with FxFG nucleoporins to translocate through the pore and import RanGDP into the nucleus.


Subject(s)
Cell Nucleus/metabolism , Nuclear Pore/metabolism , Nucleocytoplasmic Transport Proteins/chemistry , Active Transport, Cell Nucleus , Alleles , Amino Acid Sequence , Animals , Cell Survival , DNA Mutational Analysis , Dimerization , Genes, Dominant , Green Fluorescent Proteins , Immunoblotting , Luminescent Proteins/metabolism , Models, Molecular , Molecular Sequence Data , Mutagenesis, Site-Directed , Mutation , Nuclear Localization Signals , Nuclear Pore Complex Proteins/metabolism , Nucleocytoplasmic Transport Proteins/biosynthesis , Nucleocytoplasmic Transport Proteins/genetics , Phenotype , Protein Binding , Rats , Saccharomyces cerevisiae/chemistry , Saccharomyces cerevisiae/metabolism , Sequence Homology, Amino Acid , Time Factors , ran GTP-Binding Protein/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...