Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 59
Filter
1.
Cell Mol Life Sci ; 81(1): 185, 2024 Apr 17.
Article in English | MEDLINE | ID: mdl-38630271

ABSTRACT

When cells proliferate, stress on DNA replication or exposure to endogenous or external insults frequently results in DNA damage. DNA-Damage Response (DDR) networks are complex signaling pathways used by multicellular organisms to prevent DNA damage. Depending on the type of broken DNA, the various pathways, Base-Excision Repair (BER), Nucleotide Excision Repair (NER), Mismatch Repair (MMR), Homologous Recombination (HR), Non-Homologous End-Joining (NHEJ), Interstrand Crosslink (ICL) repair, and other direct repair pathways, can be activated separately or in combination to repair DNA damage. To preserve homeostasis, innate and adaptive immune responses are effective defenses against endogenous mutation or invasion by external pathogens. It is interesting to note that new research keeps showing how closely DDR components and the immune system are related. DDR and immunological response are linked by immune effectors such as the cyclic GMP-AMP synthase (cGAS)-Stimulator of Interferon Genes (STING) pathway. These effectors act as sensors of DNA damage-caused immune response. Furthermore, DDR components themselves function in immune responses to trigger the generation of inflammatory cytokines in a cascade or even trigger programmed cell death. Defective DDR components are known to disrupt genomic stability and compromise immunological responses, aggravating immune imbalance and leading to serious diseases such as cancer and autoimmune disorders. This study examines the most recent developments in the interaction between DDR elements and immunological responses. The DDR network's immune modulators' dual roles may offer new perspectives on treating infectious disorders linked to DNA damage, including cancer, and on the development of target immunotherapy.


Subject(s)
Autoimmune Diseases , Neoplasms , Humans , Adaptive Immunity , Cytokines , Apoptosis , Neoplasms/genetics
2.
Anim Nutr ; 7(4): 1087-1094, 2021 Dec.
Article in English | MEDLINE | ID: mdl-34738039

ABSTRACT

The effects of selenium (Se) yeast supplementation on performance, blood biochemical and antioxidant parameters, and milk Se content and speciation were evaluated. Thirty-six mid-lactation Holstein dairy cows were randomly assigned to 1 of 3 treatments: 1) control (basal diet containing Se at 0.11 mg/kg DM), 2) basal diet + 0.5 mg supplemental Se/kg DM (SY-0.5), and 3) basal diet + 5 mg supplemental Se/kg DM (SY-5). Selenium was supplemented as Se yeast. The trial consisted of a 1-week pretrial period and an 8-week experimental period. Milk somatic cell score decreased with SY-5 supplementation (P < 0.05), but other performance parameters were not affected (P > 0.05). The serum Se concentration increased with the increasing levels of Se yeast supplementation (P < 0.05), however, blood biochemical parameters showed few treatment effects. The antioxidant capacity of dairy cows was improved with Se yeast supplementation reflected in increased serum glutathione peroxidase activity (P < 0.05) and total antioxidant capacity (P = 0.08), and decreased malondialdehyde concentration (P < 0.05). Milk total Se concentration increased with Se dose (P < 0.05). Also, the selenomethionine concentration increased with Se dose from 13.0 ± 0.7 µg/kg in control to 33.1 ± 2.1 µg/kg in SY-0.5 and 530.4 ± 17.5 µg/kg in SY-5 cows (P < 0.05). Similarly, selenocystine concentration increased from 15.6 ± 0.9 µg/kg in control and 18.9 ± 1.1 µg/kg in SY-0.5 to 22.2 ± 1.5 µg/kg in SY-5 cows (P < 0.05). In conclusion, Se yeast is a good organic Se source to produce Se-enriched cow milk with increased Se species including selenomethionine and selenocystine. The results can provide useful information on milk Se species when a high dose Se yeast was supplemented in the cow diet.

3.
Oncol Rep ; 41(3): 1817-1826, 2019 Mar.
Article in English | MEDLINE | ID: mdl-30628703

ABSTRACT

The cancer stem cell theory recently has received enormous attention in cancer biology. Lung cancer stem­like cells are a subpopulation of undifferentiated lung tumor cells critical for lung cancer tumorigenesis, metastasis and resistance to therapy and disease relapse. The neural EGFL like 1 (NELL1) is a potent growth factor believed to preferentially target cells committed to the osteochondral lineage; yet, its expression and function in lung cancer are largely unknown. In the present study, we used specific medium to accumulate lung cancer stem­like cells of 95­D cells in spheres and obtained these highly expressed CD133 cells through flow cytometric cell sorting of CD133­stained cells which were termed 95­D lung cancer stem­like cells (95­D LCSCs). These 95­D LCSCs highly expressed stemness genes CD133, Oct4 and Sox2 determined by western blot analysis and quantitative real­time polymerase chain reaction (qPCR) analysis. Notably, we found that overexpression of NELL1 significantly reduced colony formation and invasion of 95­D LCSCs tested by soft agar colony formation and cell invasion assay. In addition, as determined by cell proliferation assay, overexpression of NELL1 increased the chemotherapeutic sensitivity of 95­D LCSCs to carboplatin and cisplatin. NELL1 also reduced the expression of phospho­MET (p­MET), Notch3 and HES1, which suggests that NELL1 may induce 95­D LCSC differentiation by inhibiting the expression of c­MET­Notch signaling. Our results suggest that NELL1 induces lung cancer stem­like cell differentiation, which provides a new potential therapeutic target for cancer stem cells.


Subject(s)
Cell Differentiation , Lung Neoplasms/pathology , Neoplastic Stem Cells/pathology , Nerve Tissue Proteins/metabolism , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Calcium-Binding Proteins , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Separation , Drug Resistance, Neoplasm , Flow Cytometry , Humans , Lung Neoplasms/drug therapy , Neoplastic Stem Cells/drug effects , Proto-Oncogene Proteins c-met/metabolism , Receptors, Notch/metabolism , Signal Transduction
4.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-792958

ABSTRACT

@# Objective: :To explore a novel chimeric antigen receptor (CAR)-T cell treatment to treat Multiple Myeloma (MM) via target B cell maturation antigen (BCMA). Methods: :A CAR-BCMA molecular was constructed based on mouse originated BCMA scFv, and was packaged into lentiviral vector and transfected into T cells from healthy donors to construct CAR-BCMA-T cells. The BCMApositive cell lines A549-BCMA, A549-BCMAOFP and K562-BCMA were constructed as target cells. Then, the CAR-BCMA-T cells were co-incubated with the constructed target cells and human myeloma U266 cells, and the cytotoxic effects of CAR-BCMA-T cells were evaluated via CCK-8 and FACS. Finally, the CAR-BCMA-T cells originated from MM patients were constructed, and its cytotoxicity against A549-BCMA were examined; in addition, the IFN-γ release level in CAR-BCMA-T cells was evaluated by ELISA and FACS. Results: After 11 days’incubation, the CAR-BCMA-T cells originated from healthy donors amplified 300 times with a positive rate of 43%. The BCMApositive target cell lines were constructed successfully. Under an effector : target ratio of 5:1, the killing rates of CARBCMA-T cells against A549-BCMA, K562-BCMA and U266 were about 80%, 60%, and 80%, respectively, which were significantly higher than those against BCMA negative cells; and the cytotoxicity was related to the BCMA expression level in target cells. What’ s more, at the effector : target ratio of 20:1, the CAR-BCMA-T cells originated from MM patients were demonstrated to exhibit a killing rate of more than 95% againstA549-BCMApositive cells, and produced large amount of IFN-γ. Conclusion: CAR-BCMA-T cells originated from both healthy and MM donors were successfully constructed, and they can effectively and specifically kill BCMA positive tumor cells.

5.
Cell Death Dis ; 9(10): 989, 2018 09 24.
Article in English | MEDLINE | ID: mdl-30250191

ABSTRACT

IL-2 is critical to the activation, growth, and survival of T cells and NK cells, and maintains the delicate balance between auto-immunity and anti-neoplasm surveillance. High IL-2 doses have clear antitumor capabilities, but also have severe side effects that limit its clinical use. Side effects include the vascular leak syndrome (VLS), which results in lung edema and liver damage. Therefore, a new version of IL-2 that does not induce organ toxicity would improve IL-2-based immunotherapy. We conducted a systematic screening by changing one amino acid at a time at the interaction area of IL-2 with its receptor IL-2R to select one particular mutant IL-2, FSD13, in which the proline at position 65 was substituted by lysine (P65L). FSD13 had a greater ability than wild-type IL-2 in stimulating CD4+ T, CD8+ T, and NK cell proliferation, enhancing the expression of CD69, CD183, CD44, and CD54 in these cells, and triggering cancer cell apoptosis. FSD13 had three-time lower than wild-type IL-2 in inducing CD4+ T to Tregs. Compared with wild-type IL-2, FSD13 greatly limited the growth, invasion into adjacent tissues, and metastasis of melanoma metastatic into the lung. In contrast to wild-type IL-2, high dose of FSD3 did not alter structures and induce any pathogenic changes in the liver and lung. Thus, we generated a novel the IL-2 mutant, FSD13, by targeting a different area than previously reported. FSD13 surpasses the wild-type IL-2's ability in stimulating the antitumor immune cell functions, but exerts much less systemic toxicity.


Subject(s)
Antineoplastic Agents/adverse effects , Drug Discovery/methods , Immunotherapy , Interleukin-2/therapeutic use , Lung Neoplasms/therapy , Melanoma, Experimental/therapy , Amino Acid Substitution , Animals , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , CD8-Positive T-Lymphocytes/metabolism , Cell Proliferation/drug effects , Coculture Techniques , Disease Models, Animal , Humans , Interleukin-2/adverse effects , Interleukin-2/genetics , K562 Cells , Killer Cells, Natural/metabolism , Lung Neoplasms/pathology , Lymphocyte Activation/drug effects , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Mutation , Treatment Outcome
6.
Article in Chinese | WPRIM (Western Pacific) | ID: wpr-821285

ABSTRACT

@#[Abstract] Objective: To establish a chimeric antigen receptor(CAR)modified T cells specifically targeting CD19 molecule (CD19CAR-T cells) and to testify their in vitro killing effect on target cells. Methods: CD19-CAR fragments yielded by PCR were constructed into pCDH-GFP lentiviral vectors by molecular cloning technology. The packaged lentiviral particles were transducted into CD3+ T cells of donors. Transduction efficiency was measured by flow cytometry and PCR. The in vitro cytotoxicity of obtained CD19CAR-T cells against CD19+ Ramos cells was tested by 7-AAD staining. Results: The amplification folds of CD3+ T cells increased to (78.8± 23.2) folds after in vitro culture for 10 days, and about (58.3±5.4)% cells expressing GFP.About (57.4±9.3)% CD19+Ramos cells were specifically killed by the CD19-CAR-T cells in vitro at the E∶T ratio of 5∶1. Conclusion: This study successfully established an effective method for constructing and amplifying CD19-CAR-T cells in vitro, which showed profound efficiency and specific cytotoxity against CD19+ Ramos cells.And this report might provide an experimental evidence for clinical treatment of CD19+ B cell neoplasmas.

7.
Oncotarget ; 8(32): 53302-53312, 2017 Aug 08.
Article in English | MEDLINE | ID: mdl-28881812

ABSTRACT

Many studies are based on the hypothesis that recurrence and drug resistance in lung carcinoma are due to a subpopulation of cancer stem-like cells (CSLCs) in solid tumors. Therefore it is crucial to screen for and recognize lung CSLCs. In this study, we stimulated non-small cell lung cancer (NSCLC) A549 cells to display stem cell-like characteristics using a combination of five small molecule compounds. The putative A549 stem cells activated an important CSLC marker, CD133 protein, as well multiple CSLC-related genes including ATP-binding cassette transporter G2 (ABCG2), C-X-C chemokine receptor type 4 (CXCR4), NESTIN, and BMI1. The A549 stem-like cells displayed resistance to the chemotherapeutic drugs etoposide and cisplatin, epithelial-to-mesenchymal transition properties, and increased protein expression levels of NOTCH1 and Hes Family bHLH Transcription Factor 1 (HES1). When A549 cells were pretreated with a NOTCH signaling pathway inhibitor before compound induction, expression of the NOTCH1 target gene HES1 was reduced. This demonstrated that the NOTCH signaling pathway in the putative A549 stem-like cells had been activated. Together, the results of our study showed that a combination of five small molecule agents could transform A549 cells into putative stem-like cells, and that these compounds could also elevate CD133 and ABCG2 protein expression levels in H460 cells. This study provides a convenient method for obtaining lung CSLCs, which may be an effective strategy for developing lung carcinoma treatments.

8.
Toxicol Sci ; 158(2): 275-285, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28482072

ABSTRACT

Endoplasmic reticulum (ER) stress has been implicated in the development of maternal diabetes-induced neural tube defects (NTDs). ER stress-induced C/EBP homologous protein (CHOP) plays an important role in the pro-apoptotic execution pathways. However, the molecular mechanism underlying ER stress- and CHOP-induced neuroepithelium cell apoptosis in diabetic embryopathy is still unclear. Deletion of the Chop gene significantly reduced maternal diabetes-induced NTDs. CHOP deficiency abrogated maternal diabetes-induced mitochondrial dysfunction and neuroepithelium cell apoptosis. Further analysis demonstrated that CHOP repressed the expression of peroxisome-proliferator-activated receptor-γ coactivator-1α (PGC-1α), an essential regulator for mitochondrial biogenesis and function. Both CHOP deficiency in vivo and knockdown in vitro restore high glucose-suppressed PGC-1α expression. In contrast, CHOP overexpression mimicked inhibition of PGC-1α by high glucose. In response to the ER stress inducer tunicamycin, PGC-1α expression was decreased, whereas the ER stress inhibitor 4-phenylbutyric acid blocked high glucose-suppressed PGC-1α expression. Moreover, maternal diabetes in vivo and high glucose in vitro promoted the interaction between CHOP and the PGC-1α transcriptional regulator CCAAT/enhancer binding protein-ß (C/EBPß), and reduced C/EBPß binding to the PGC-1α promoter leading to markedly decrease in PGC-1α expression. Together, our findings support the hypothesis that maternal diabetes-induced ER stress increases CHOP expression which represses PGC-1α through suppressing the C/EBPß transcriptional activity, subsequently induces mitochondrial dysfunction and ultimately results in NTDs.


Subject(s)
Diabetes, Gestational/physiopathology , Endoplasmic Reticulum Stress/physiology , Fetal Diseases/physiopathology , Mitochondria/physiology , Peroxisome Proliferator-Activated Receptor Gamma Coactivator 1-alpha/antagonists & inhibitors , Transcription Factor CHOP/physiology , Animals , Apoptosis/genetics , Cell Line , Dimerization , Female , Gene Expression Regulation/drug effects , Glucose/administration & dosage , Mice , Mice, Inbred C57BL , Neural Tube Defects/genetics , Pregnancy , Transcription Factor CHOP/genetics , Tunicamycin/pharmacology
9.
Oncogene ; 35(38): 5056-5069, 2016 09 22.
Article in English | MEDLINE | ID: mdl-27041584

ABSTRACT

Melanoma is one of the most lethal forms of skin cancer because of its early metastatic spread. The variant form of CD44 (CD44v), a cell surface glycoprotein, is highly expressed on metastatic melanoma. The mechanisms of regulation of CD44 alternative splicing in melanoma and its pathogenic contributions are so far poorly understood. Here, we investigated the expression level of CD44 in a large set of melanocytic lesions at different stages. We found that the expression of CD44v8-10 and a splicing factor, U2AF2, is significantly increased during melanoma progression, whereas CD82/KAI1, a tetraspanin family of tumor suppressor, is reduced in metastatic melanoma. CD44v8-10 and U2AF2 expression levels, which are negatively correlated with CD82 levels, are markedly elevated in primary melanoma compared with dysplastic nevi and further increased in metastatic melanoma. We also showed that patients with higher CD44v8-10 and U2AF2 expression levels tended to have shorter survival. By using both in vivo and in vitro assays, we demonstrated that CD82 inhibits the production of CD44v8-10 on melanoma. Mechanistically, U2AF2 is a downstream target of CD82 and in malignant melanoma facilitates CD44v8-10 alternative splicing. U2AF2-mediated CD44 isoform switch is required for melanoma migration in vitro and lung and liver metastasis in vivo. Notably, overexpression of CD82 suppresses U2AF2 activity by inducing U2AF2 ubiquitination. In addition, our data suggested that enhancement of melanoma migration by U2AF2-dependent CD44v8-10 splicing is mediated by Src/focal adhesion kinase/RhoA activation and formation of stress fibers, as well as CD44-E-selectin binding reinforcement. These findings uncovered a hitherto unappreciated function of CD82 in severing the linkage between U2AF2-mediated CD44 alternative splicing and cancer aggressiveness, with potential prognostic and therapeutic implications in melanoma.


Subject(s)
Hyaluronan Receptors/genetics , Kangai-1 Protein/genetics , Melanoma/genetics , Splicing Factor U2AF/genetics , Alternative Splicing/genetics , Animals , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Melanocytes/pathology , Melanoma/pathology , Mice , Neoplasm Metastasis , Phosphorylation , Prognosis , Protein Isoforms/genetics , Proteolysis , Ubiquitination/genetics , Xenograft Model Antitumor Assays
10.
ACS Med Chem Lett ; 7(4): 397-402, 2016 Apr 14.
Article in English | MEDLINE | ID: mdl-27096048

ABSTRACT

Structure-activity relationship exploration of the historical biarylurea series led to the identification of novel CNS penetrant CXCR2 antagonists with nanomolar potency, favorable PK profile, and good developability potentials. More importantly, the key compound 22 showed efficacy in a cuprizone-induced demyelination model with twice daily oral administration, thereby supporting CXCR2 to be a potential therapeutic target for the treatment of demyelinating diseases such as multiple sclerosis.

11.
J Biol Chem ; 291(5): 2087-106, 2016 Jan 29.
Article in English | MEDLINE | ID: mdl-26504080

ABSTRACT

Tumor invasiveness depends on the ability of tumor cells to breach endothelial barriers. In this study, we investigated the mechanism by which the adhesion of melanoma cells to endothelium regulates adherens junction integrity and modulates tumor transendothelial migration (TEM) by initiating thrombin generation. We found that the B-Raf(V600E) mutation in metastatic melanoma cells up-regulated tissue factor (TF) expression on cell membranes and promoted thrombin production. Co-culture of endothelial monolayers with metastatic melanoma cells mediated the opening of inter-endothelial spaces near melanoma cell contact sites in the presence of platelet-free plasma (PFP). By using small interfering RNA (siRNA), we demonstrated that B-Raf(V600E) and TF silencing attenuated the focal disassembly of adherens junction induced by tumor contact. Vascular endothelial-cadherin (VE-cadherin) disassembly was dependent on phosphorylation of p120-catenin on Ser-879 and VE-cadherin on Tyr-658, Tyr-685, and Tyr-731, which can be prevented by treatment with the thrombin inhibitor, hirudin, or by silencing the thrombin receptor, protease-activated receptor-1, in endothelial cells. We also provided strong evidence that tumor-derived thrombin enhanced melanoma TEM by inducing ubiquitination-coupled VE-cadherin internalization, focal adhesion formation, and actin assembly in endothelium. Confocal microscopic analysis of tumor TEM revealed that junctions transiently opened and resealed as tumor cells accomplished TEM. In addition, in the presence of PFP, tumor cells preferentially transmigrated via paracellular routes. PFP supported melanoma transmigration under shear conditions via a B-Raf(V600E)-thrombin-dependent mechanism. We concluded that the activation of thrombin generation by cancer cells in plasma is an important process regulating melanoma extravasation by disrupting endothelial junction integrity.


Subject(s)
Endothelium, Vascular/metabolism , Melanoma/metabolism , Proto-Oncogene Proteins B-raf/metabolism , Skin Neoplasms/metabolism , Thrombin/metabolism , Thromboplastin/metabolism , Antigens, CD/metabolism , Cadherins/metabolism , Cell Adhesion , Cell Line, Tumor , Cell Membrane/metabolism , Cell Movement , Electric Impedance , Gene Silencing , Human Umbilical Vein Endothelial Cells , Humans , Mutation , Neoplasm Metastasis , Permeability , Phosphorylation , Plasma/metabolism , RNA, Small Interfering/metabolism , Ubiquitination
12.
Toxicol Lett ; 234(3): 151-61, 2015 May 05.
Article in English | MEDLINE | ID: mdl-25725129

ABSTRACT

Diphenyl difluoroketone (EF24), a curcumin analog, exhibits potent anti-tumor activities by arresting cell cycle and inducing apoptosis. However, the efficacy and modes of action of EF24 on melanoma metastasis remain elusive. In this study, we found that at non-cytotoxic concentrations, EF24 suppressed cell motility and epithelial-to-mesenchymal Transition (EMT) of melanoma cell lines, Lu1205 and A375. EF24 also suppressed HMGA2 expression at mRNA and protein levels. miR-33b directly bound to HMGA2 3' untranslated region (3'-UTR) to suppress its expression as measured by dual-luciferase assay. EF24 increased expression of E-cadherin and decreased STAT3 phosphorylation and expression of the mesenchymal markers, vimentin and N-cadherin. miR-33b inhibition or HMGA2 overexpression reverted EF24-mediated suppression of EMT phenotypes. In addition, EF24 modulated the HMGA2-dependent actin stress fiber formation, focal adhesion assembly and FAK, Src and RhoA activation by targeting miR-33b. Thus, the results suggest that EF24 suppresses melanoma metastasis via upregulating miR-33b and concomitantly reducing HMGA2 expression. The observed activities of EF24 support its further evaluation as an anti-metastatic agent in melanoma therapy.


Subject(s)
Antineoplastic Agents/pharmacology , Benzylidene Compounds/pharmacology , Cell Movement/physiology , Curcumin/analogs & derivatives , Epithelial-Mesenchymal Transition/drug effects , HMGA2 Protein/drug effects , Melanoma/metabolism , MicroRNAs/drug effects , Piperidones/pharmacology , Blotting, Western , Cell Line, Tumor , Cell Movement/drug effects , Epithelial-Mesenchymal Transition/physiology , Fluorescent Antibody Technique , Humans , Melanoma/drug therapy , Melanoma/physiopathology , MicroRNAs/physiology , Reverse Transcriptase Polymerase Chain Reaction , Up-Regulation/drug effects
13.
Lung Cancer ; 87(2): 98-106, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25512094

ABSTRACT

BACKGROUND: Since cancer stem cells exhibit embryonic-like self-renewal characteristics and malignant behavior, including drug resistance and metastasis, they may be the origin of tumorigenesis and cancer recurrence. Cancer cell stemness is also highly relevant to cancer in hypoxic environments. METHODS: In our study, we used cobalt dichloride (CoCl2) to create a hypoxic environment for lung adenocarcinoma A549 cells and the cisplatine-resistant cell line A549/DDP. The cancer stem-like CD166 positive population and the cells' stemness were detected by flowcytometry and quantitative real-time PCR after separation using magnetic antibodies. Drug resistance to cisplatine, docetaxel and pemetrexed was also measured. Finally, a tissue array was used to analyze the relationship between hypoxia-induced stemness and overall survival after radical surgery. RESULTS: Data showed that chemical-induced hypoxia changed cell stemness by enhancing stem cell transcription factors and markers of chemotherapeutic drug resistance. The CD166-positive cancer stem cell-like population showed greater drug resistance than the CD166-negative cells. Tissue array studies also suggested a poorer prognosis for patients whose tissue expressed higher CD166 levels. CONCLUSION: Our findings indicate that chemical hypoxia may augment cancer cell stemness and drug resistance in CD166-positive stem cells. Therefore, targeting the stem-like cell population, especially CD166-positive cells, may represent a novel therapeutic strategy to treat lung cancer.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/mortality , Drug Resistance, Neoplasm/genetics , Lung Neoplasms/genetics , Lung Neoplasms/mortality , Neoplastic Stem Cells/metabolism , Activated-Leukocyte Cell Adhesion Molecule/genetics , Activated-Leukocyte Cell Adhesion Molecule/metabolism , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Adenocarcinoma of Lung , Adult , Aged , Aged, 80 and over , Antineoplastic Agents/pharmacology , Biomarkers , Cell Hypoxia/genetics , Cell Line, Tumor , Dose-Response Relationship, Drug , Female , Gene Expression , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Immunophenotyping , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Grading , Neoplasm Staging , Prognosis , RNA, Messenger/genetics , RNA, Messenger/metabolism , Survival Analysis , Young Adult
14.
Stem Cells Transl Med ; 3(3): 356-64, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24375541

ABSTRACT

Targeting cancer stem cells (CSCs) with immunotherapy may be an effective means to prevent recurrences in glioblastoma multiforme (GBM). It is well established that CD133 is expressed in the population of GBM tumor cells representing CSCs. This raises a possibility that CD133 could serve as a potential target for cytotoxic T cells (CTLs) to target glioblastoma cancer stem cells. Two potential human leukocyte antigen (HLA)-A*0201-restricted CD133 epitopes, ILSAFSVYV (CD133-405) and YLQWIEFSI (CD133-753), showed strong binding to HLA-A*0201 molecules. In vitro immunogenicity studies generated peptide-specific CD8(+) CTLs from normal donors. Autologous monocyte-derived dendritic cells pulsed with the CD133-405 or CD133-753 peptides generated CTLs that efficiently recognized the CD133 epitopes presented in T2 HLA-A*0201 cells and specifically lysed CD133+ HLA-A*0201(+) GBM CSCs. These studies demonstrated natural processing and subsequent presentation of these epitopes in GBM CSCs and the ability of CTLs to kill CSCs bearing the antigen. Immunization studies in mice using the mouse homolog CD133 epitopes demonstrated immunogenicity in the absence of autoimmune damage. The results presented in this study support the use of CD133-specific epitope vaccines to target CSCs in glioblastoma and other cancers.


Subject(s)
Antigens, CD/immunology , Central Nervous System Neoplasms/therapy , Epitopes, T-Lymphocyte/immunology , Glioblastoma/therapy , Glycoproteins/immunology , HLA-A2 Antigen/immunology , Immunotherapy/methods , Neoplastic Stem Cells/immunology , Peptides/immunology , T-Lymphocytes, Cytotoxic/immunology , AC133 Antigen , Animals , Antigens, CD/genetics , Cell- and Tissue-Based Therapy , Central Nervous System Neoplasms/genetics , Central Nervous System Neoplasms/immunology , Central Nervous System Neoplasms/pathology , Cytotoxicity, Immunologic , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/immunology , Epitopes, T-Lymphocyte/genetics , Gene Expression/immunology , Glioblastoma/genetics , Glioblastoma/immunology , Glioblastoma/pathology , Glycoproteins/genetics , HLA-A2 Antigen/genetics , Humans , Mice , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/pathology , Oligopeptides/chemical synthesis , Oligopeptides/immunology , Oligopeptides/pharmacology , Peptides/genetics , Protein Binding , Secondary Prevention , T-Lymphocytes, Cytotoxic/cytology , T-Lymphocytes, Cytotoxic/drug effects
15.
PLoS One ; 8(8): e70192, 2013.
Article in English | MEDLINE | ID: mdl-23936390

ABSTRACT

Our pilot study using miRNA arrays found that miRNA-29c (miR-29c) is differentially expressed in the paired low-metastatic lung cancer cell line 95C compared to the high-metastatic lung cancer cell line 95D. Bioinformatics analysis shows that integrin ß1 and matrix metalloproteinase 2 (MMP2) could be important target genes of miR-29c. Therefore, we hypothesized that miR-29c suppresses lung cancer cell adhesion to extracellular matrix (ECM) and metastasis by targeting integrin ß1 and MMP2. The gain-of-function studies that raised miR-29c expression in 95D cells by using its mimics showed reductions in cell proliferation, adhesion to ECM, invasion and migration. In contrasts, loss-of-function studies that reduced miR-29c by using its inhibitor in 95C cells promoted proliferation, adhesion to ECM, invasion and migration. Furthermore, the dual-luciferase reporter assay demonstrated that miR-29c inhibited the expression of the luciferase gene containing the 3'-UTRs of integrin ß1 and MMP2 mRNA. Western blotting indicated that miR-29c downregulated the expression of integrin ß1 and MMP2 at the protein level. Gelatin zymography analysis further confirmed that miR-29c decreased MMP2 enzyme activity. Nude mice with xenograft models of lung cancer cells confirmed that miR-29c inhibited lung cancer metastasis in vivo, including bone and liver metastasis. Taken together, our results demonstrate that miR-29c serves as a tumor metastasis suppressor, which suppresses lung cancer cell adhesion to ECM and metastasis by directly inhibiting integrin ß1 and MMP2 expression and by further reducing MMP2 enzyme activity. The results show that miR-29c may be a novel therapeutic candidate target to slow lung cancer metastasis.


Subject(s)
Extracellular Matrix/pathology , Integrin beta1/genetics , Lung Neoplasms/pathology , Matrix Metalloproteinase 1/genetics , MicroRNAs/genetics , 3' Untranslated Regions/genetics , Animals , Base Sequence , Cattle , Cell Adhesion/genetics , Cell Line, Tumor , Cell Proliferation , Down-Regulation/genetics , Female , Lung Neoplasms/genetics , Mice , Mice, Nude , Neoplasm Invasiveness , Neoplasm Metastasis , Rats
16.
Prostate ; 73(13): 1378-90, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23728788

ABSTRACT

BACKGROUND: Prostate cancer stem cells (PCSC) offer theoretical explanations to many clinical and biological behaviors of the disease in human. In contrast to approaches of using side populations and cell-surface markers to isolate and characterize the putative PCSC, we hypothesize that androgen deprivation leads to functional enrichment of putative PCSC. METHODS AND RESULTS: Human prostate cancer lines LNCaP, LAPC4 and LAPC9 were depleted of androgen in cell cultures and in castrated SCID mice. The resultant androgen deprivation-resistant or castration-resistant populations, in particular in LNCaP and its derivative cell lines, displayed increased expression of pluripotency transactivators and significantly higher tumorigenicity. Individual tumor cell clones were isolated from castration-resistant bulk cultures of LNCaP (CR-LNCaP) and tested for tumorigenicity in male SCID mice under limiting dilution conditions. As few as 200 cells were able to form spheres in vitro, and generate tumors with similar growth kinetics as 10(6) LNCaP or 10(4) CR-LNCaP cells in vivo. These putative PCSC were CD44(+) /CD24(-) and lack the expression of prostate lineage proteins. When transplanted into the prostate of an intact male SCID mouse, these putative PCSC seemed to show limited differentiation into Ck5(+) , Ck8(+) , Ck5(+) /Ck8(+) , and AR(+) cells. On the other hand, stable transduction of LNCaP with retrovirus encoding Sox2 led to androgen-deprivation resistant growth and down-regulation of major prostate lineage gene products in vitro. CONCLUSION: Concurrence of overexpression of pluripotency transactivators and resistance to androgen deprivation supported the role of putative PCSC in the emergence of prostate cancer resistant to androgen deprivation.


Subject(s)
Androgens/metabolism , Neoplastic Stem Cells/pathology , Prostate/pathology , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Up-Regulation , Animals , Castration , Cell Line, Tumor , Gene Expression Regulation, Neoplastic , Humans , Male , Mice , Mice, SCID , Neoplastic Stem Cells/metabolism , Prostate/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Receptors, Androgen/genetics
17.
Toxicology ; 308: 104-12, 2013 Jun 07.
Article in English | MEDLINE | ID: mdl-23562787

ABSTRACT

Integrin ß1 facilitates cancer cell adhesion, migration and metastasis by activating intracellular signaling pathways including the ERK and PI3K signaling pathways. In previous studies, shikonin, an active naphthoquinone isolated from the Chinese medicine Zi Cao (gromwell), showed effective anticancer activity both in vivo and in vitro. However, the mechanisms underlying shikonin's anticancer activity are not fully elucidated. Increasing evidence indicates that shikonin inhibits tumor metastasis, but little is known about the effect of shikonin on lung cancer cells. To better understand the anti-metastatic role of shikonin in lung cancer, in this study we sought to investigate the effect of shikonin on lung cancer cell proliferation, adhesion to extracellular matrices (ECM), migration and invasion in non-small cell lung cancer A549 cells. We also sought to investigate the molecular mechanisms underlying shikonin's anticancer effects. Here we showed that when non-small cell lung cancer A549 cells were treated with shikonin for 24h, 8.0µM shikonin significantly inhibited cell proliferation, while cells treated with less than 2.0µM shikonin for 24h significantly suppressed cell adhesion to the ECM, invasion and migration in a dose-dependent manner. Moreover, real-time PCR and Western blot analysis showed that shikonin led to a reduction in the expression of integrin ß1 at the mRNA and protein levels. Further elucidation of the mechanisms involved revealed that shikonin repressed the phosphorylation of extracellular signal-regulated kinase (ERK1/2). Taken together, our findings provide new evidence that shikonin suppresses lung cancer invasion and metastasis by inhibiting integrin ß1 expression and the ERK1/2 signaling pathway.


Subject(s)
Carcinoma, Non-Small-Cell Lung/metabolism , Extracellular Matrix/metabolism , Integrin beta1/metabolism , Lung Neoplasms/metabolism , MAP Kinase Signaling System/physiology , Naphthoquinones/pharmacology , Antineoplastic Agents, Phytogenic/pharmacology , Antineoplastic Agents, Phytogenic/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Cell Adhesion/drug effects , Cell Adhesion/physiology , Cell Line, Tumor , Extracellular Matrix/drug effects , Gene Expression Regulation, Neoplastic/drug effects , Humans , Integrin beta1/biosynthesis , Lung Neoplasms/drug therapy , Lung Neoplasms/pathology , MAP Kinase Signaling System/drug effects , Naphthoquinones/therapeutic use
18.
Oncol Rep ; 29(6): 2181-90, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23563700

ABSTRACT

Lung cancer is still the leading cause of cancer-related deaths worldwide. Identifying new oncogenic drivers and developing efficient inhibitors through molecular targeting approaches are crucial for improving therapies. The aim of this study was to investigate whether targeting fibroblast growth factor receptor 1 (FGFR1) with ponatinib inhibits the cell growth in both established and primary lung cancer cells overexpressing FGFR1. Eighty-eight non-small cell lung cancer (NSCLC) and paired normal tissue specimens were analyzed by real-time RT-PCR for FGFR1 gene expression. We identified four cell lines and two newly established primary lung cancer cultures that showed high FGFR1 expression levels, and evaluated the effect of the novel FGFR1 inhibitor ponatinib on cell growth. Approximately 50% (30 out of 59) NSCLC specimens expressed FGFR1>2-fold compared with their adjacent normal counterparts using quantitative RT-PCR. Ponatinib treatment of established NSCLC cell lines expressing higher levels of FGFR1 resulted in marked cell growth inhibition and suppression of clonogenicity. This growth inhibition was associated with inactivation of FGFR1 and its downstream targets. FGFR1 knockdown by shRNA achieved similar results when compared to treatment with ponatinib. Furthermore, ponatinib was able to significantly inhibit the growth of primary lung cancer cultures in vitro. Our data indicate that pharmacological inhibition of FGFR1 kinase activity with ponatinib may be effective for the treatment of lung cancer patients whose tumors overexpress FGFR1.


Subject(s)
Antineoplastic Agents/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Imidazoles/pharmacology , Lung Neoplasms/drug therapy , Pyridazines/pharmacology , Receptor, Fibroblast Growth Factor, Type 1/metabolism , Adult , Aged , Carcinoma, Non-Small-Cell Lung/metabolism , Cell Proliferation/drug effects , Female , Gene Expression , Gene Knockdown Techniques , Humans , Lung Neoplasms/metabolism , Male , Middle Aged , Molecular Targeted Therapy , Phosphorylation , Protein Processing, Post-Translational/drug effects , Receptor, Fibroblast Growth Factor, Type 1/antagonists & inhibitors , Receptor, Fibroblast Growth Factor, Type 1/genetics , Signal Transduction/drug effects , Tumor Cells, Cultured/drug effects
19.
Toxicology ; 303: 139-46, 2013 Jan 07.
Article in English | MEDLINE | ID: mdl-23146760

ABSTRACT

Epithelial-to-mesenchymal transition (EMT) is a cellular process during which epithelial polarized cells become motile mesenchymal-appearing cells, which in turn promotes carcinoma invasion and metastasis. Resveratrol (trans-3,4',5-trihydroxystilbene) is a natural polyphenolic compound found in grapes, red wine and several other plants. Numerous reports in the literature indicate that resveratrol can suppress cancer invasion and metastasis. However, the underlying mechanisms of inhibiting metastasis by resveratrol are complex, not fully elucidated and the subject of intense scientific debate. Despite evidence indicating that EMT can be a target for resveratrol, little is known about the effect of resveratrol on lung cancer cells. Our previous studies demonstrated that TGF-ß1 induces EMT to promote lung adenocarcinoma invasion and metastasis. To understand the repressive role of resveratrol in lung cancer invasion and metastasis, we sought to investigate the potential use of resveratrol as an inhibitor of TGF-ß1-induced EMT development in A549 lung cancer cells in vitro. Here we show that when A549 cells are treated with TGF-ß1 and resveratrol, the latter inhibits the initiation of TGF-ß1-induced EMT. Our results show that 20 µM resveratrol increases expression of the epithelial phenotype marker E-cadherin and represses the expression of the mesenchymal phenotype markers, Fibronectin and Vimentin during the initiation of TGF-ß1-induced EMT. Resveratrol also inhibits expression of EMT-inducing transcription factors Snail1 and Slug, although the expression of the Twist1 transcription factor remained unchanged. Resveratrol inhibits the TGF-ß1-induced increase in cell adhesion, migration and invasion of A549 lung cancer cells. Taken together, our findings provide new evidence that resveratrol suppresses lung cancer invasion and metastasis in vitro through inhibiting TGF-ß1-induced EMT.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Epithelial-Mesenchymal Transition/drug effects , Lung Neoplasms/drug therapy , Stilbenes/pharmacology , Antineoplastic Agents, Phytogenic/administration & dosage , Cadherins/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Fibronectins/genetics , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lung Neoplasms/pathology , Neoplasm Invasiveness , Neoplasm Metastasis , Resveratrol , Stilbenes/administration & dosage , Transforming Growth Factor beta1/pharmacology , Vimentin/genetics
20.
Mol Cancer Ther ; 8(9): 2746-53, 2009 Sep.
Article in English | MEDLINE | ID: mdl-19723878

ABSTRACT

Malignant gliomas manifest frequent tumor recurrence after surgical resection and/or other treatment because of their nature of invasiveness and dissemination. The recognized brain tumor-tracking property of neural progenitor/stem cells opened the possibility of targeting malignant brain tumors using neural progenitor/stem cells. We and others have previously shown that fetal neural progenitor/stem cells can be used to deliver therapeutic molecules to brain tumors. Our recent work has further shown that gene delivery by bone marrow-derived neural progenitor/stem cells achieves therapeutic effects in a glioma model. In this study, we isolate and characterize bone marrow-derived neural progenitor/stem cells, which also express the chemokine receptor chemokine CXC receptor 4 (CXCR4). We show that CXCR4 is required for their chemotaxis and extracellular matrix invasion against a gradient of glioma soluble factors. Furthermore, beta-galactosidase-labeled bone marrow-derived neural progenitor/stem cells implanted in the contralateral side of the brain were shown to track gliomas as early as day 1 and increased through days 3 and 7. Intracranial glioma tracking by bone marrow-derived neural progenitor/stem cells is significantly inhibited by preincubation of bone marrow-derived neural progenitor/stem cells with a blocking anti-CXCR4 antibody, suggesting a CXCR4-dependent tracking mechanism. Glioma tracking bone marrow-derived neural progenitor/stem cells were found to express progenitor/stem cell markers, as well as CXCR4. Although bromodeoxyuridine incorporation assays and proliferating antigen staining indicated that tumor tracking bone marrow-derived neural progenitor/stem cells were mostly nonproliferating, these cells survive in the local tumor environment with little apoptosis. Elucidating the molecular mechanism of brain tumor tracking by adult source stem cells may provide basis for the development of future targeted therapy for malignant brain tumors.


Subject(s)
Bone Marrow Cells/metabolism , Brain Neoplasms/metabolism , Glioma/metabolism , Neurons/metabolism , Receptors, CXCR4/metabolism , Stem Cells/metabolism , Animals , Cell Line, Tumor , Enzyme-Linked Immunosorbent Assay , Immunohistochemistry , Rats , Rats, Inbred F344
SELECTION OF CITATIONS
SEARCH DETAIL
...