Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
Add more filters










Publication year range
1.
Elife ; 82019 05 30.
Article in English | MEDLINE | ID: mdl-31144617

ABSTRACT

Squamous cell carcinomas (SCCs) account for the majority of cancer mortalities. Although TP63 is an established lineage-survival oncogene in SCCs, therapeutic strategies have not been developed to target TP63 or it's downstream effectors. In this study we demonstrate that TP63 directly regulates NRG1 expression in human SCC cell lines and that NRG1 is a critical component of the TP63 transcriptional program. Notably, we show that squamous tumors are dependent NRG1 signaling in vivo, in both genetically engineered mouse models and human xenograft models, and demonstrate that inhibition of NRG1 induces keratinization and terminal squamous differentiation of tumor cells, blocking proliferation and inhibiting tumor growth. Together, our findings identify a lineage-specific function of NRG1 in SCCs of diverse anatomic origin.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Carcinoma, Squamous Cell/pathology , Cell Differentiation , Neuregulin-1/metabolism , Transcription Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Carcinoma, Squamous Cell/genetics , Cell Line, Tumor , Cell Proliferation , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, Nude , Receptor, ErbB-3/metabolism
2.
Sci Rep ; 8(1): 8239, 2018 05 29.
Article in English | MEDLINE | ID: mdl-29844389

ABSTRACT

Neuregulin 1 (NRG1) is required for development of the central and peripheral nervous system and regulates neurotransmission in the adult. NRG1 and the gene encoding its receptor, ERBB4, are risk genes for schizophrenia, although how alterations in these genes disrupt their function has not been fully established. Studies of knockout and transgenic mice have yielded conflicting results, with both gain and loss of function resulting in similar behavioral and electrophysiological phenotypes. Here, we used high affinity antibodies to NRG1 and ErbB4 to perturb the function of the endogenous proteins in adult mice. Treatment with NRG1 antibodies that block receptor binding caused behavioral alterations associated with schizophrenia, including, hyper-locomotion and impaired pre-pulse inhibition of startle (PPI). Electrophysiological analysis of brain slices from anti-NRG1 treated mice revealed reduced synaptic transmission and enhanced paired-pulse facilitation. In contrast, mice treated with more potent ErbB4 function blocking antibodies did not display behavioral alterations, suggesting a receptor independent mechanism of the anti-NRG1-induced phenotypes. We demonstrate that anti-NRG1 causes accumulation of the full-length transmembrane protein and increases phospho-cofilin levels, which has previously been linked to impaired synaptic transmission, indicating enhancement of non-canonical NRG1 signaling could mediate the CNS effects.


Subject(s)
Central Nervous System/physiology , Electrophysiology/methods , Neuregulin-1/metabolism , Schizophrenia/metabolism , Actin Depolymerizing Factors/metabolism , Animals , Antibodies, Blocking/administration & dosage , Disease Models, Animal , Genetic Predisposition to Disease , HEK293 Cells , Humans , Mice , Mice, Inbred C57BL , Neuregulin-1/genetics , Neuregulin-1/immunology , Protein Stability , Receptor, ErbB-4/genetics , Receptor, ErbB-4/immunology , Receptor, ErbB-4/metabolism , Risk , Schizophrenia/genetics , Signal Transduction , Synaptic Transmission
3.
Cancer Cell ; 32(2): 221-237.e13, 2017 08 14.
Article in English | MEDLINE | ID: mdl-28781121

ABSTRACT

Maintenance of phenotypic heterogeneity within cell populations is an evolutionarily conserved mechanism that underlies population survival upon stressful exposures. We show that the genomes of a cancer cell subpopulation that survives treatment with otherwise lethal drugs, the drug-tolerant persisters (DTPs), exhibit a repressed chromatin state characterized by increased methylation of histone H3 lysines 9 and 27 (H3K9 and H3K27). We also show that survival of DTPs is, in part, maintained by regulators of H3K9me3-mediated heterochromatin formation and that the observed increase in H3K9me3 in DTPs is most prominent over long interspersed repeat element 1 (LINE-1). Disruption of the repressive chromatin over LINE-1 elements in DTPs results in DTP ablation, which is partially rescued by reducing LINE-1 expression or function.


Subject(s)
Chromatin/genetics , Drug Resistance, Neoplasm/genetics , Epigenetic Repression/drug effects , Long Interspersed Nucleotide Elements/genetics , Neoplasms/pathology , Animals , Antineoplastic Agents/pharmacology , Gene Expression Regulation, Neoplastic , Genomic Instability/drug effects , Histone-Lysine N-Methyltransferase/metabolism , Histones/metabolism , Humans , Methylation , Mice , Mice, Nude , Mice, SCID , Neoplasms/drug therapy , Neoplasms/genetics , Stress, Physiological , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
4.
Nat Chem Biol ; 12(7): 531-8, 2016 07.
Article in English | MEDLINE | ID: mdl-27214401

ABSTRACT

The KDM5 family of histone demethylases catalyzes the demethylation of histone H3 on lysine 4 (H3K4) and is required for the survival of drug-tolerant persister cancer cells (DTPs). Here we report the discovery and characterization of the specific KDM5 inhibitor CPI-455. The crystal structure of KDM5A revealed the mechanism of inhibition of CPI-455 as well as the topological arrangements of protein domains that influence substrate binding. CPI-455 mediated KDM5 inhibition, elevated global levels of H3K4 trimethylation (H3K4me3) and decreased the number of DTPs in multiple cancer cell line models treated with standard chemotherapy or targeted agents. These findings show that pretreatment of cancer cells with a KDM5-specific inhibitor results in the ablation of a subpopulation of cancer cells that can serve as the founders for therapeutic relapse.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Neoplasm/drug effects , Enzyme Inhibitors/pharmacology , Neoplasms/drug therapy , Neoplasms/pathology , Retinoblastoma-Binding Protein 2/antagonists & inhibitors , Antineoplastic Agents/chemistry , Cell Survival/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemistry , Humans , Models, Molecular , Molecular Structure , Retinoblastoma-Binding Protein 2/metabolism , Structure-Activity Relationship
5.
Development ; 141(21): 4087-97, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25336737

ABSTRACT

In vertebrate retina, histogenesis occurs over an extended period. To sustain the temporal generation of diverse cell types, retinal progenitor cells (RPCs) must self-renew. However, self-renewal and regulation of RPCs remain poorly understood. Here, we demonstrate that cell-extrinsic factors coordinate with the epigenetic regulator high-mobility group AT-hook 2 (Hmga2) to regulate self-renewal of late retinal progenitor cells (RPCs). We observed that a small subset of RPCs was capable of clonal propagation and retained multipotentiality of parents in the presence of endothelial cells (ECs), known self-renewal regulators in various stem cell niches. The self-renewing effects, also observed in vivo, involve multiple intercellular signaling pathways, engaging Hmga2. As progenitors exhaust during retinal development, expression of Hmga2 progressively decreases. Analyses of Hmga2-expression perturbation, in vitro and in vivo, revealed that Hmga2 functionally helps to mediate cell-extrinsic influences on late-retinal progenitor self-renewal. Our results provide a framework for integrating the diverse intercellular influences elicited by epigenetic regulators for self-renewal in a dynamic stem cell niche: the developing vertebrate retina.


Subject(s)
HMGA2 Protein/metabolism , Retina/cytology , Retina/metabolism , Stem Cells/cytology , Stem Cells/metabolism , Animals , Cell Cycle/genetics , Cell Cycle/physiology , Cell Differentiation/genetics , Cell Differentiation/physiology , Cell Proliferation , Cells, Cultured , Female , HMGA2 Protein/genetics , Pregnancy , Rats , Rats, Sprague-Dawley , Signal Transduction/genetics , Signal Transduction/physiology
6.
PLoS One ; 8(8): e70352, 2013.
Article in English | MEDLINE | ID: mdl-23936412

ABSTRACT

Earlier, we reported that CTLA4 expression is inversely correlated with CD38 expression in chronic lymphocytic leukemia (CLL) cells. However, the specific role of CTLA4 in CLL pathogenesis remains unknown. Therefore, to elucidate the possible role of CTLA4 in CLL pathogenesis, CTLA4 was down-regulated in primary CLL cells. We then evaluated proliferation/survival in these cells using MTT, (3)H-thymidine uptake and Annexin-V apoptosis assays. We also measured expression levels of downstream molecules involved in B-cell proliferation/survival signaling including STAT1, NFATC2, c-Fos, c-Myc, and Bcl-2 using microarray, PCR, western blotting analyses, and a stromal cell culture system. CLL cells with CTLA4 down-regulation demonstrated a significant increase in proliferation and survival along with an increased expression of STAT1, STAT1 phosphorylation, NFATC2, c-Fos phosphorylation, c-Myc, Ki-67 and Bcl-2 molecules. In addition, compared to controls, the CTLA4-downregulated CLL cells showed a decreased frequency of apoptosis, which also correlated with increased expression of Bcl-2. Interestingly, CLL cells from lymph node and CLL cells co-cultured on stroma expressed lower levels of CTLA4 and higher levels of c-Fos, c-Myc, and Bcl-2 compared to CLL control cells. These results indicate that microenvironment-controlled-CTLA4 expression mediates proliferation/survival of CLL cells by regulating the expression/activation of STAT1, NFATC2, c-Fos, c-Myc, and/or Bcl-2.


Subject(s)
CTLA-4 Antigen/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , ADP-ribosyl Cyclase 1/metabolism , Apoptosis , B-Lymphocytes/pathology , CTLA-4 Antigen/deficiency , CTLA-4 Antigen/genetics , Cell Proliferation , Cell Survival , Down-Regulation/genetics , Gene Silencing , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Prognosis , RNA, Small Interfering/genetics , Tumor Microenvironment
7.
Sci Transl Med ; 5(171): 171ra18, 2013 Feb 06.
Article in English | MEDLINE | ID: mdl-23390248

ABSTRACT

Although standard chemotherapies are commonly used to treat most types of solid tumors, such treatment often results in inadequate response to, or relapse after, therapy. This is particularly relevant for lung cancer because most patients are diagnosed with advanced-stage disease and are treated with frontline chemotherapy. By studying the residual tumor cells that remain after chemotherapy in several in vivo non-small cell lung cancer models, we found that these cells have increased levels of human epidermal growth factor receptor (HER) signaling due, in part, to the enrichment of a preexisting NRG1(HI) subpopulation. Neuregulin 1 (NRG1) signaling in these models can be mediated by either the HER3 or HER4 receptor, resulting in the differential activation of downstream effectors. Inhibition of NRG1 signaling inhibits primary tumor growth and enhances the magnitude and duration of the response to chemotherapy. Moreover, we show that inhibition of ligand-mediated Her4 signaling impedes disease relapse in cases where NRG1 inhibition is insufficient. These findings demonstrate that ligand-dependent Her4 signaling plays an important role in disease relapse.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , ErbB Receptors/metabolism , Lung Neoplasms/drug therapy , Neuregulin-1/antagonists & inhibitors , Signal Transduction , Animals , Antibodies, Blocking/pharmacology , Antibodies, Blocking/therapeutic use , Autocrine Communication/drug effects , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , ErbB Receptors/antagonists & inhibitors , Humans , Ligands , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , Mice , Mice, Nude , Neoplasm, Residual/drug therapy , Neoplasm, Residual/metabolism , Neoplasm, Residual/pathology , Neuregulin-1/metabolism , Receptor, ErbB-4 , Signal Transduction/drug effects , Treatment Outcome , Xenograft Model Antitumor Assays
8.
PLoS One ; 7(10): e45647, 2012.
Article in English | MEDLINE | ID: mdl-23115623

ABSTRACT

Although chemotherapy is used to treat most advanced solid tumors, recurrent disease is still the major cause of cancer-related mortality. Cancer stem cells (CSCs) have been the focus of intense research in recent years because they provide a possible explanation for disease relapse. However, the precise role of CSCs in recurrent disease remains poorly understood and surprisingly little attention has been focused on studying the cells responsible for re-initiating tumor growth within the original host after chemotherapy treatment. We utilized both xenograft and genetically engineered mouse models of non-small cell lung cancer (NSCLC) to characterize the residual tumor cells that survive chemotherapy treatment and go on to cause tumor regrowth, which we refer to as tumor re-initiating cells (TRICs). We set out to determine whether TRICs display characteristics of CSCs, and whether assays used to define CSCs also provide an accurate readout of a cell's ability to cause tumor recurrence. We did not find consistent enrichment of CSC marker positive cells or enhanced tumor initiating potential in TRICs. However, TRICs from all models do appear to be in EMT, a state that has been linked to chemoresistance in numerous types of cancer. Thus, the standard CSC assays may not accurately reflect a cell's ability to drive disease recurrence.


Subject(s)
Antineoplastic Agents/therapeutic use , Carcinoma, Non-Small-Cell Lung/drug therapy , Lung Neoplasms/drug therapy , Animals , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Disease Models, Animal , Epithelial-Mesenchymal Transition , Flow Cytometry , Humans , Lung Neoplasms/pathology , Mice , Neoplastic Stem Cells/pathology , Xenograft Model Antitumor Assays
9.
Int J Cancer ; 131(12): 2951-60, 2012 Dec 15.
Article in English | MEDLINE | ID: mdl-22511234

ABSTRACT

Mantle cell lymphoma (MCL) is one of the most aggressive B-cell lymphomas with a median patient survival of only 5-7 years. The failure of existing therapies is mainly due to disease relapse when therapy-resistant tumor cells remain after chemotherapy. Therefore, development and testing of novel therapeutic strategies to target these therapy-resistant MCL are needed. Here, we developed an in vivo model of therapy-resistant MCL by transplanting a patient-derived MCL cell line (Granta 519) into NOD/SCID mice followed by treatment with combination chemotherapy. Cytomorphologic, immunophenotypic, in vitro and in vivo growth analyses of these therapy-resistant MCL cells confirm their MCL origin and resistance to chemotherapy. Moreover, quantitative real-time PCR revealed the upregulation of GLI transcription factors, which are mediators of the hedgehog signaling pathway, in these therapy-resistant MCL cells. Therefore, we developed an effective therapeutic strategy for resistant MCL by treating the NOD/SCID mice bearing Granta 519 MCL with CHOP chemotherapy to reduce tumor burden combined with GLI-antisense oligonucleotides or bortezomib, a proteosome inhibitor, to target therapy-resistant MCL cells that remained after chemotherapy. This regimen was followed by treatment with MCL-specific cytotoxic T lymphocytes to eliminate all detectable leftover minimal residual disease. Mice treated with this strategy showed a significantly increased survival and decreased tumor burden compared to the mice in all other groups. Such therapeutic strategies that combine chemotherapy with targeted therapy followed by tumor-specific immunotherapy are effective and have excellent potential for clinical application to provide long-term, disease-free survival in MCL patients.


Subject(s)
Hedgehog Proteins/metabolism , Lymphoma, Mantle-Cell/drug therapy , Signal Transduction , Animals , Antineoplastic Agents/therapeutic use , Humans , Mice , Mice, Inbred NOD , Mice, SCID , Real-Time Polymerase Chain Reaction
10.
Cancer Immunol Immunother ; 61(10): 1819-32, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22441656

ABSTRACT

Mantle cell lymphoma (MCL) is an aggressive B-cell neoplasm with few patients achieving long-term survival with current treatment regimens. High-dose therapy is effective in reducing the tumor burden; however, patients eventually relapse due to minimal residual disease. Having demonstrated efficacy in other malignancies, the effectiveness of dendritic cell-based immunotherapy for minimal residual MCL was examined. We demonstrated that dendritic cells (DC) primed with MCL antigens stimulated the activation of MCL-specific T cells that recognized and destroyed both MCL cell lines and primary MCL in vitro. In addition, in vivo studies demonstrated that adoptively transferred MCL-specific T cells were able to significantly inhibit tumor growth in mice with minimal residual MCL. Subsequently, when combined with CHOP chemotherapy, adoptive T-cell therapy was able to significantly extend the survival of the mice by further reducing the tumor burden. These results clearly show that MCL-specific cellular immunotherapy is effective in treating minimal residual MCL, paving the way for future clinical studies.


Subject(s)
Immunotherapy, Adoptive/methods , Kidney Neoplasms/therapy , Liver Neoplasms/therapy , Lung Neoplasms/therapy , Lymphoma, Mantle-Cell/therapy , T-Lymphocytes/transplantation , Animals , Antigens, Neoplasm/immunology , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Cell Line, Tumor , Combined Modality Therapy , Cyclophosphamide/therapeutic use , Dendritic Cells/immunology , Doxorubicin/therapeutic use , Humans , Kidney Neoplasms/drug therapy , Kidney Neoplasms/immunology , Kidney Neoplasms/mortality , Kidney Neoplasms/secondary , Liver Neoplasms/drug therapy , Liver Neoplasms/immunology , Liver Neoplasms/mortality , Liver Neoplasms/secondary , Lung Neoplasms/drug therapy , Lung Neoplasms/immunology , Lung Neoplasms/mortality , Lung Neoplasms/secondary , Lymphocyte Activation/immunology , Lymphoma, Mantle-Cell/drug therapy , Lymphoma, Mantle-Cell/immunology , Lymphoma, Mantle-Cell/mortality , Lymphoma, Mantle-Cell/pathology , Mice , Mice, Inbred NOD , Neoplasm, Residual , Prednisone/therapeutic use , T-Lymphocytes/immunology , Treatment Outcome , Vincristine/therapeutic use
11.
Mol Cancer Res ; 6(12): 1928-36, 2008 Dec.
Article in English | MEDLINE | ID: mdl-19074837

ABSTRACT

B-cell chronic lymphocytic leukemia (B-CLL) is characterized by an accumulation of neoplastic B cells due to their resistance to apoptosis and increased survival. Among various factors, the tumor microenvironment is known to play a role in the regulation of cell proliferation and survival of many cancers. However, it remains unclear how the tumor microenvironment contributes to the increased survival of B-CLL cells. Therefore, we studied the influence of bone marrow stromal cell-induced hedgehog (Hh) signaling on the survival of B-CLL cells. Our results show that a Hh signaling inhibitor, cyclopamine, inhibits bone marrow stromal cell-induced survival of B-CLL cells, suggesting a role for Hh signaling in the survival of B-CLL cells. Furthermore, gene expression profiling of primary B-CLL cells (n = 48) indicates that the expression of Hh signaling molecules, such as GLI1, GLI2, SUFU, and BCL2, is significantly increased and correlates with disease progression of B-CLL patients with clinical outcome. In addition, SUFU and GLI1 transcripts, as determined by real-time PCR, are significantly overexpressed and correlate with adverse indicators of clinical outcome in B-CLL patients, such as cytogenetics or CD38 expression. Furthermore, selective down-regulation of GLI1 by antisense oligodeoxynucleotides (GLI1-ASO) results in decreased BCL2 expression and cell survival, suggesting that GLI1 may regulate BCL2 and, thereby, modulate cell survival in B-CLL. In addition, there was significantly increased apoptosis of B-CLL cells when cultured in the presence of GLI1-ASO and fludarabine. Together, these results reveal that Hh signaling is important in the pathogenesis of B-CLL and, hence, may be a potential therapeutic target.


Subject(s)
Hedgehog Proteins/metabolism , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Leukemia, Lymphocytic, Chronic, B-Cell/physiopathology , Stromal Cells/cytology , Stromal Cells/physiology , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Apoptosis/physiology , Cell Communication/drug effects , Cell Communication/physiology , Cell Division/drug effects , Cell Division/physiology , Cell Survival/drug effects , Cell Survival/physiology , Gene Expression Profiling , Gene Expression Regulation, Leukemic , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/drug therapy , Oligodeoxyribonucleotides, Antisense/pharmacology , Signal Transduction/drug effects , Signal Transduction/physiology , Transcription Factors/genetics , Vidarabine/analogs & derivatives , Vidarabine/pharmacology , Zinc Finger Protein GLI1
12.
Dev Neurosci ; 30(6): 389-409, 2008.
Article in English | MEDLINE | ID: mdl-19033687

ABSTRACT

The canonical Wnt pathway is known to influence multiple developmental events such as patterning, cell proliferation and cell specification. Recent studies have provided evidence of the involvement of the canonical Wnt pathway in the emergence and development of the optic neuroepithelium and its derivatives, particularly the retina. However, the mechanism of its action during retinal development remains rather obscure. Here, we demonstrate that (in agreement with observations in the blood, intestine, and skin) the canonical Wnt pathway influences retinal development by maintaining stem cells/progenitors. For example, the activation of this pathway keeps the early retinal stem cells/progenitors proliferating and uncommitted, while its attenuation facilitates their differentiation into retinal ganglion cells in vitro and in vivo. In addition, we demonstrate that Wnt signaling acts in concert with Notch signaling during retinal histogenesis, where the latter calibrates the influence of the former on the differentiation status of retinal stem cells/progenitors by regulating Lef1 and sFRP2.


Subject(s)
Receptors, Notch/metabolism , Retina/cytology , Retina/metabolism , Signal Transduction/physiology , Stem Cells/physiology , Wnt Proteins/metabolism , Animals , Cell Differentiation/physiology , Cell Proliferation , Chick Embryo , Female , Lymphoid Enhancer-Binding Factor 1/genetics , Lymphoid Enhancer-Binding Factor 1/metabolism , Oligonucleotide Array Sequence Analysis , Pregnancy , Rats , Rats, Sprague-Dawley , Receptors, Notch/genetics , Retina/embryology , Stem Cells/cytology , Wnt Proteins/genetics
13.
Mol Cancer Ther ; 7(6): 1450-60, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18524848

ABSTRACT

Mantle cell lymphoma (MCL) has one of the worst clinical outcomes among the B-cell lymphomas, with a median survival of only 3 to 4 years. Therefore, a better understanding of the underlying mechanisms that regulate MCL proliferation/survival is needed to develop an effective therapy. Because sonic hedgehog (Shh)-GLI signaling has been shown to be important in the proliferation and survival of several cancers, and no such information is available for MCL, this study was undertaken. Our results show that the molecules associated with Shh-GLI signaling, such as PTCH and SMO receptors, and GLI1 and GLI2 target transcription factors were expressed in the human MCL cell lines and primary MCL cells from patients. Perturbation of this signaling in the presence of exogenous Shh/cyclopamine significantly (P < 0.001) influenced the proliferation of JVM2 MCL cells. Furthermore, down-regulation of GLI transcription factors using antisense oligonucleotides not only resulted in significantly (P < 0.001) decreased proliferation of the MCL cells but also significantly (P < 0.05) increased their susceptibility to chemotherapeutic drug, doxorubicin. Also, down-regulation of GLI decreased cyclin D1 and BCL2 transcript levels, which suggests that these key molecules might be regulated by GLI in MCL. Thus, our results indicate a significant role for Shh-GLI signaling in the proliferation of MCL, and molecular targeting of GLI is a potential therapeutic approach to improve the treatment for MCL.


Subject(s)
Hedgehog Proteins/metabolism , Lymphoma, Mantle-Cell/therapy , Signal Transduction , Transcription Factors/metabolism , Cell Line, Tumor , Cell Proliferation/drug effects , Cyclin D1/genetics , Cyclin D1/metabolism , Down-Regulation/drug effects , Doxorubicin/pharmacology , Gene Expression Regulation, Neoplastic/genetics , Hedgehog Proteins/genetics , Humans , Lymphoma, Mantle-Cell/genetics , Lymphoma, Mantle-Cell/pathology , Models, Biological , Proto-Oncogene Proteins c-bcl-2/genetics , Proto-Oncogene Proteins c-bcl-2/metabolism , Signal Transduction/drug effects , Transcription Factors/genetics , Zinc Finger Protein GLI1
14.
Int Immunopharmacol ; 8(6): 819-27, 2008 Jun.
Article in English | MEDLINE | ID: mdl-18442785

ABSTRACT

A partial mechanism by which a conformationally-biased, response-selective agonist of complement component C5a, Tyr-Ser-Phe-Lys-Pro-Met-Pro-Leu-D-Ala-Arg or YSFKPMPLaR (EP54), acts as a molecular adjuvant is presented by showing the manner in which this peptide engages human dendritic cells (DC). Confocal microscopy was used to show that fluorescent-labeled EP54 (0.2 microM) and fluorescent-labeled B and T cell epitopes attached to EP54 (i.e., EP54-containing vaccines, 0.2 microM) were internalized by human DCs well within 30 min of exposure. After 24 h of exposure, EP54 and the B and T cell epitopes of the EP54-containing vaccines (20 microM) were presented on the DC surface in the context of HLA-ABC and HLA-DR determinants. Also, exposure of DCs to EP54 (50 microg/ml) induced the activation of genes specific for the Th1 cytokines IL-6, IL-12, INFgamma, and TNFalpha as well as the Th2 cytokine IL-4. Internalization, HLA expression, and cytokine gene activation were not observed in the presence of the inactive, scrambled EP54 constructs arguing that these effects of EP54 are mediated predominately via C5a receptors on the DC surface.


Subject(s)
Antigen Presentation , Complement C5a/agonists , Dendritic Cells/immunology , Peptide Fragments/immunology , Adjuvants, Immunologic , Complement C5a/immunology , Complement C5a/pharmacology , Cytokines/immunology , Cytokines/metabolism , Dendritic Cells/cytology , Dendritic Cells/drug effects , Dendritic Cells/metabolism , Epitopes, B-Lymphocyte/immunology , Epitopes, B-Lymphocyte/metabolism , Epitopes, T-Lymphocyte/immunology , Epitopes, T-Lymphocyte/metabolism , HLA-DR Antigens/immunology , Histocompatibility Antigens Class I/immunology , Humans , Peptide Fragments/pharmacology
15.
J Neuroimmune Pharmacol ; 2(2): 202-12, 2007 Jun.
Article in English | MEDLINE | ID: mdl-18040845

ABSTRACT

Tumors of the nervous system, including neuroblastoma and glioblastoma, are difficult to treat with current therapies. Despite the advances in cancer therapeutics, the outcomes in these patients remain poor and, therefore, new modalities are required. Recent literature demonstrates that cytotoxic effector cells can effectively kill tumors of the nervous system. In addition, we have previously shown that umbilical cord blood (UCB) contains precursors of antitumor cytotoxic effector cells. Therefore, to evaluate the antitumor potential of UCB-derived effector cells, studies were designed to compare the in vitro and in vivo antitumor effects of UCB- and peripheral blood (PB)-derived antigen-nonspecific and antigen-specific effector cells against tumors of the nervous system. Mononuclear cells (MNCs) from UCB were used to generate both interleukin-2 (IL-2)-activated killer (LAK) cells and tumor-specific cytotoxic T lymphocytes (CTLs). UCB-derived LAK cells showed a significant in vitro cytotoxicity against IMR-32, SK-NMC, and U-87 human neuroblastoma and glioblastoma, respectively. In addition, the CTLs generated using dendritic cells primed with IMR-32 tumor cell lysate showed a selective cytotoxicity in vitro against IMR-32 cells, but not against U-87 or MDA-231 cells. Furthermore, treatment of SCID mice bearing IMR-32 neuroblastoma with tumor-specific CTLs resulted in a significant (p < 0.01) inhibition of tumor growth and increased overall survival. Thus, these results demonstrate the potential of UCB-derived effector cells against human neuroblastoma and warrant further preclinical studies.


Subject(s)
Fetal Blood/cytology , Fetal Blood/immunology , Immunotherapy/methods , Neuroblastoma/immunology , Neuroblastoma/therapy , Animals , Cell Line, Tumor , Cells, Cultured , Cytotoxicity, Immunologic/immunology , Fetal Blood/transplantation , Glioblastoma/immunology , Glioblastoma/therapy , Humans , Killer Cells, Lymphokine-Activated/immunology , Killer Cells, Lymphokine-Activated/transplantation , Leukocytes, Mononuclear/immunology , Leukocytes, Mononuclear/transplantation , Mice , Mice, SCID , T-Lymphocytes, Cytotoxic/immunology , T-Lymphocytes, Cytotoxic/transplantation
16.
J Cardiovasc Pharmacol Ther ; 12(3): 237-47, 2007 Sep.
Article in English | MEDLINE | ID: mdl-17875952

ABSTRACT

A therapeutic strategy that would mitigate the events leading to hyperplasia and facilitate re-endothelialization of an injured artery after balloon angioplasty could be effective for a long-term patency of the artery. It is hypothesized that erythropoietin (EPO), which has both anti-inflammatory and antiapoptotic properties, will prevent hyperplasia, and its ability to proliferate and mobilize endothelial progenitor cells will re-endothelialize the injured artery. To test this hypothesis, EPO (5000 IU/kg) in solution was injected intraperitoneally 6 hours before vascular injury and then on every alternate day for a week or as a single dose (5000 IU/kg) in a sustained release gel formulation 1 week before the vascular injury. Morphometric analysis revealed nearly continuous re-endothelialization of the injured artery in EPO solution-treated animals (90% vs less than 20% in saline control); however, the treatment also caused excessive neointima formation (intima/media ratio, 2.10 +/- 0.09 vs 1.60 +/- 0.02 saline control, n = 5, P < .001). The EPO gel also induced similar excessive neointima formation. Immunohistochemical analysis of the injured arteries from the animals treated with EPO solution demonstrated a significant angiogenic response in adventitia and media, thus explaining the formation of excessive neointima. Although the results are in contrast to expectation, they explain a greater degree of stenosis seen in hemodialysis access fistulas in patients who are on EPO therapy for anemic condition. The results also caution the use of EPO, particularly in patients who are at a risk of vascular injury or are suffering from an atherosclerotic condition.


Subject(s)
Carotid Artery Injuries/drug therapy , Erythropoietin/pharmacology , Tunica Intima/drug effects , Animals , Carotid Arteries/pathology , Cell Movement , Cell Proliferation/drug effects , Delayed-Action Preparations , Disease Models, Animal , Endothelial Cells/drug effects , Endothelium, Vascular/drug effects , Erythropoietin/administration & dosage , Erythropoietin/adverse effects , Gels , Hyperplasia/chemically induced , Immunohistochemistry , Injections, Intraperitoneal , Neovascularization, Pathologic/chemically induced , Rats , Rats, Sprague-Dawley , Stem Cells/drug effects , Stem Cells/metabolism , Tunica Intima/pathology
17.
J Biol Chem ; 282(48): 35187-201, 2007 Nov 30.
Article in English | MEDLINE | ID: mdl-17855369

ABSTRACT

Based on a variety of approaches, evidence suggests that different cell types in the vertebrate retina are generated by multipotential progenitors in response to interactions between cell intrinsic and cell extrinsic factors. The identity of some of the cellular determinants that mediate such interactions has emerged, shedding light on mechanisms underlying cell differentiation. For example, we know now that Notch signaling mediates the influence of the microenvironment on states of commitment of the progenitors by activating transcriptional repressors. Cell intrinsic factors such as the proneural basic helix-loop-helix and homeodomain transcription factors regulate a network of genes necessary for cell differentiation and maturation. What is missing from this picture is the role of developmental chromatin remodeling in coordinating the expression of disparate classes of genes for the differentiation of retinal progenitors. Here we describe the role of Brm, an ATPase in the SWI/SNF chromatin remodeling complex, in the differentiation of retinal progenitors into retinal ganglion cells. Using the perturbation of expression and function analyses, we demonstrate that Brm promotes retinal ganglion cell differentiation by facilitating the expression and function of a key regulator of retinal ganglion cells, Brn3b, and the inhibition of Notch signaling. In addition, we demonstrate that Brm promotes cell cycle exit during retinal ganglion cell differentiation. Together, our results suggest that Brm represents one of the nexus where diverse information of cell differentiation is integrated during cell differentiation.


Subject(s)
Adenosine Triphosphatases/physiology , Cell Cycle Proteins/physiology , Gene Expression Regulation, Developmental , Gene Expression Regulation , Receptor, Notch1/biosynthesis , Retina/embryology , Transcription Factor Brn-3B/biosynthesis , Transcription Factors/metabolism , Adenosine Triphosphatases/genetics , Animals , Cell Cycle Proteins/genetics , Cell Differentiation , Cell Lineage , Cell Proliferation , Chromatin/metabolism , DNA Helicases , Models, Biological , Nuclear Proteins , Rats , Rats, Sprague-Dawley , Retina/cytology , Signal Transduction , Stem Cells/cytology , Transcription, Genetic
18.
Clin Cancer Res ; 13(18 Pt 1): 5295-304, 2007 Sep 15.
Article in English | MEDLINE | ID: mdl-17875758

ABSTRACT

PURPOSE: In B-cell chronic lymphocytic leukemia (CLL), high CD38 expression has been associated with unfavorable clinical course, advanced disease, resistance to therapy, shorter time to first treatment, and shorter survival. However, the genes associated with CLL patient subgroups with high and low CD38 expression and their potential role in disease progression is not known. EXPERIMENTAL DESIGN: To identify the genes associated with the clinical disparity in CLL patients with high versus low CD38 expression, transcriptional profiles were obtained from CLL cells from 39 different patients using oligonucleotide microarray. Gene expression was also compared between CLL cells and B cells from healthy individuals. RESULTS: Gene expression analysis identified 76 differentially expressed genes in CD38 high versus low groups. Out of these genes, HEM1, CTLA4, and MNDA were selected for further studies and their differential expression was confirmed by real-time PCR. HEM1 overexpression was associated with poor outcome, whereas the overexpression of CTLA4 and MNDA was associated with good outcome. Down-regulation of HEM1 expression in patient CLL cells resulted in a significant increase in their susceptibility to fludarabine-mediated killing. In addition, when gene expression patterns in CD38 high and low CLL cells were compared with normal B-cell profiles, ATM expression was found to be significantly lower in CD38 high compared with CD38 low CLL as confirmed by real-time reverse transcription-PCR. CONCLUSIONS: These results identify the possible genes that may be involved in cell proliferation and survival and, thus, determining the clinical behavior of CLL patients expressing high or low CD38.


Subject(s)
ADP-ribosyl Cyclase 1/genetics , Gene Expression Regulation, Leukemic , Genes, Neoplasm , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/mortality , Antigens, CD/genetics , Antigens, Differentiation/genetics , Antigens, Differentiation, Myelomonocytic/genetics , Ataxia Telangiectasia Mutated Proteins , CTLA-4 Antigen , Cell Cycle Proteins/genetics , Cell Proliferation , DNA-Binding Proteins/genetics , Disease Progression , Gene Expression Profiling , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Membrane Proteins/genetics , Prognosis , Protein Serine-Threonine Kinases/genetics , Transcription Factors/genetics , Tumor Suppressor Proteins/genetics
19.
Int J Mol Med ; 20(4): 461-9, 2007 Oct.
Article in English | MEDLINE | ID: mdl-17786276

ABSTRACT

In B-cell chronic lymphocytic leukemia (CLL), Rai stage, immunoglobulin gene mutational status, chromosomal abnormalities, CD38 and ZAP-70 expression were used as prognostic markers. In this study, to understand the molecular basis of chromosomal abnormalities leading to tumor progression, 90 CLL patients were grouped into poor prognosis (with 11q deletion and trisomy 12) and good prognosis (with normal karyotype and 13q deletion) and their clinical outcome was assessed. Gene expression profiles of 35 CLL samples with poor outcome (11q deletion, n=9; trisomy 12, n=5) and good outcome (13q deletion, n=13; normal karyotype, n=8) were analyzed using oligonucleotide microarray. Significance analysis of microarray (SAM) identified 27 differentially expressed genes between these two subgroups with significant overexpression of ATF5 and underexpression of CDC16, PCDH8, SLAM, MNDA and ATF2 in CLL patients with poor outcome. ATF5 gene expression in CLL was further studied because of its role in the regulation of cell cycle progression/differentiation and apoptosis. The overexpression of ATF5 was confirmed by real-time PCR using 39 CLL samples from the poor and good outcome groups. ATF5 was significantly (p<0.001) overexpressed in the poor outcome group. Furthermore, ATF5 expression was significantly higher in the 11q deletion as well as trisomy 12 group alone compared to the 13q deletion and normal karyotype groups. ATF5 overexpression was also associated with significantly (p=0.04) shorter time to treatment. Similarly, expression of five underexpressed genes also correlated with longer time to treatment. Thus, this report demonstrates that ATF5 may be one of the key genes involved in increased proliferation and survival in 11q deletion or trisomy 12, whereas CD16, CD86, SLAM, MNDA and ATF2 may be involved in the decreased proliferation of CLL cells with 13q deletion or normal karyotype.


Subject(s)
Chromosome Deletion , Chromosomes, Human, Pair 11/genetics , Chromosomes, Human, Pair 12/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Trisomy/genetics , Activating Transcription Factors/metabolism , Cluster Analysis , Cytogenetic Analysis , Disease Progression , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genes, Neoplasm , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/therapy , Male , Middle Aged , Prognosis , Time Factors , Treatment Outcome
20.
Cancer Genet Cytogenet ; 172(2): 120-6, 2007 Jan 15.
Article in English | MEDLINE | ID: mdl-17213020

ABSTRACT

B-cell chronic lymphocytic leukemia (B-CLL) is the most common B-cell leukemia among older populations in Western countries. The clinical course of B-CLL is heterogeneous: in some patients the disease course is indolent, in others it is aggressive. The B-CLL subgroups with chromosome 11q23 deletion have been associated with aggressive disease course involving ATM deletion, extensive bulky lymphadenopathy (BLA), and inferior clinical outcome. Using real-time reverse transcriptase-polymerase chain reaction, we found that ATM was consistently underexpressed in B-CLL patients with BLA, irrespective of 11q23 deletion status. In addition, B-CLL patients who presented with BLA had a significantly shorter time to treatment (2 months) than did patients without BLA (74 months). Moreover, gene expression analysis in B-CLL patients with and without BLA revealed differences in expression for genes involved in apoptosis, cell cycle, and cell adhesion. These results indicate an association between BLA and reduced expression of ATM, suggesting a role for ATM in disease progression in B-CLL.


Subject(s)
Cell Cycle Proteins/biosynthesis , Cell Cycle Proteins/genetics , Chromosome Deletion , Chromosomes, Human, Pair 11/genetics , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , Down-Regulation/genetics , Gene Expression Regulation, Leukemic , Leukemia, Lymphocytic, Chronic, B-Cell/genetics , Lymphatic Diseases/genetics , Protein Serine-Threonine Kinases/biosynthesis , Protein Serine-Threonine Kinases/genetics , Tumor Suppressor Proteins/biosynthesis , Tumor Suppressor Proteins/genetics , Ataxia Telangiectasia Mutated Proteins , Cell Adhesion/genetics , Cell Cycle/genetics , Cell Cycle Proteins/antagonists & inhibitors , DNA-Binding Proteins/antagonists & inhibitors , Female , Humans , Leukemia, Lymphocytic, Chronic, B-Cell/diagnosis , Leukemia, Lymphocytic, Chronic, B-Cell/pathology , Lymphatic Diseases/diagnosis , Lymphatic Diseases/pathology , Male , Middle Aged , Prognosis , Protein Serine-Threonine Kinases/antagonists & inhibitors , Tumor Suppressor Proteins/antagonists & inhibitors
SELECTION OF CITATIONS
SEARCH DETAIL
...