Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 15 de 15
Filter
1.
Front Immunol ; 14: 1186422, 2023.
Article in English | MEDLINE | ID: mdl-37441074

ABSTRACT

Introduction: A multiple organ dysfunction syndrome (MODS) workshop convened by the National Institute of Child Health and Human Development in 2015 identified acute respiratory distress syndrome (ARDS) and complications of allogeneic blood and marrow transplantation (allo-BMT) as contributors to MODS in pediatric patients. Pulmonary dysfunction also remains a significant complication of allo-BMT. Idiopathic pneumonia syndrome (IPS) defines non-infectious, acute, lung injury that occurs post-transplant. Injury and activation to endothelial cells (ECs) contribute to each form of lung inflammation. Methods: Two murine models were employed. In an ARDS model, naïve B6 mice receive an intravenous (i.v.) injection of lipopolysaccharide (LPS). In the established model of IPS, naïve B6D2F1 mice receive lethal total body irradiation followed by BMT from either allogeneic (B6) or syngeneic (B6D2F1) donors. Lung inflammation was subsequently assessed in each scenario. Results: Intravenous injection of LPS to B6 mice resulted in enhanced mRNA expression of TNFα, IL-6, Ang-2, E-, and P-selectin in whole lung homogenates. The expression of Ang-2 in this context is regulated in part by TNFα. Additionally, EC activation was associated with increased total protein and cellularity in broncho-alveolar lavage fluid (BALF). Similar findings were noted during the development of experimental IPS. We hypothesized that interventions maintaining EC integrity would reduce the severity of ARDS and IPS. Defibrotide (DF) is FDA approved for the treatment of BMT patients with sinusoidal obstruction syndrome and renal or pulmonary dysfunction. DF stabilizes activated ECs and protect them from further injury. Intravenous administration of DF before and after LPS injection significantly reduced mRNA expression of TNFα, IL6, Ang-2, E-, and P-selectin compared to controls. BALF showed decreased cellularity, reflecting less EC damage and leak. Allogeneic BMT mice were treated from day -1 through day 14 with DF intraperitoneally, and lungs were harvested at 3 weeks. Compared to controls, DF treatment reduced mRNA expression of TNFα, IL6, Ang-2, E-, and P- selectin, BALF cellularity, and lung histopathology. Conclusion: The administration of DF modulates EC injury in models of ARDS and IPS. Cytokine inhibition in combination with agents that stabilize EC integrity may be an attractive strategy for patients in each setting.


Subject(s)
Acute Lung Injury , Pneumonia , Respiratory Distress Syndrome , Humans , Mice , Animals , Child , Lipopolysaccharides/toxicity , Tumor Necrosis Factor-alpha/genetics , P-Selectin , Endothelial Cells/pathology , Interleukin-6/genetics , Lung/pathology , Pneumonia/etiology , Pneumonia/prevention & control , Pneumonia/pathology , Acute Lung Injury/etiology , Acute Lung Injury/therapy , Respiratory Distress Syndrome/therapy , Respiratory Distress Syndrome/complications , RNA, Messenger
2.
J Clin Invest ; 132(13)2022 07 01.
Article in English | MEDLINE | ID: mdl-35608905

ABSTRACT

DNA methyltransferase 3a (DNMT3a) is an important part of the epigenetic machinery that stabilizes patterns of activated T cell responses. We hypothesized that donor T cell DNMT3a regulates alloreactivity after allogeneic blood and marrow transplantation (allo-BMT). T cell conditional Dnmt3a KO mice were used as donors in allo-BMT models. Mice receiving allo-BMT from KO donors developed severe acute graft-versus-host disease (aGVHD), with increases in inflammatory cytokine levels and organ histopathology scores. KO T cells migrated and proliferated in secondary lymphoid organs earlier and demonstrated an advantage in trafficking to the small intestine. Donor T cell subsets were purified after BMT for whole-genome bisulfite sequencing (WGBS) and RNA-Seq. KO T cells had global methylation similar to that of WT cells, with distinct, localized areas of hypomethylation. Using a highly sensitive computational method, we produced a comprehensive profile of the altered epigenome landscape. Hypomethylation corresponded with changes in gene expression in several pathways of T cell signaling and differentiation. Additionally, Dnmt3a-KO T cells resulted in superior graft-versus-tumor activity. Our findings demonstrate a critical role for DNMT3a in regulating T cell alloreactivity and reveal pathways that control T cell tolerance. These results also provide a platform for deciphering clinical data that associate donor DNMT3a mutations with increased GVHD, decreased relapse, and improved survival.


Subject(s)
Graft vs Host Disease , Hematopoietic Stem Cell Transplantation , Animals , Bone Marrow Transplantation/methods , Graft vs Host Disease/genetics , Mice , T-Lymphocytes , Transplantation, Homologous/methods
3.
Methods Mol Biol ; 2255: 159-169, 2021.
Article in English | MEDLINE | ID: mdl-34033102

ABSTRACT

Cytotoxic T cell-induced cell death is well documented. Cytotoxic T cell releases various cytolytic proteins. The cytolytic proteins induce target cell death. T cell-induced cell death can be measured by the lytic assay. One of the well-known lytic assays uses radioactive tracer, Chromium-51 (51Cr), and detects the amount of 51Cr released from target cells. This assay can detect cell death and the efficiency of the T cell-induced cell death by coculture effector cells (T cells) and target cells. This assay can determine the kinetics of the cell lysis. The issue of this approach is the use of radioactive material. This chapter describes measuring T cell-induced cell death by determining the epigenetic remodeling and the release of cytolytic proteins. Determine the efficiency of T cell-induced cell death by using a flow cytometry-based detection method.


Subject(s)
CD8-Positive T-Lymphocytes/immunology , Chromium Radioisotopes/analysis , Colonic Neoplasms/pathology , Cytotoxicity Tests, Immunologic/methods , Flow Cytometry/methods , Lymphoma, B-Cell/pathology , T-Lymphocytes, Cytotoxic/immunology , Adenocarcinoma/immunology , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Cell Death , Colonic Neoplasms/immunology , Colonic Neoplasms/metabolism , Lymphoma, B-Cell/immunology , Lymphoma, B-Cell/metabolism , Mice , Tumor Cells, Cultured
4.
Immunity ; 39(1): 184-95, 2013 Jul 25.
Article in English | MEDLINE | ID: mdl-23890071

ABSTRACT

The study of T cell memory and the target of vaccine design have focused on memory subsumed by T cells bearing the αß T cell receptor. Alternatively, γδ T cells are thought to provide rapid immunity, particularly at mucosal borders. Here, we have shown that a distinct subset of mucosal γδ T cells mounts an immune response to oral Listeria monocytogenes (Lm) infection and leads to the development of multifunctional memory T cells capable of simultaneously producing interferon-γ and interleukin-17A in the murine intestinal mucosa. Challenge infection with oral Lm, but not oral Salmonella or intravenous Lm, induced rapid expansion of memory γδ T cells, suggesting contextual specificity to the priming pathogen. Importantly, memory γδ T cells were able to provide enhanced protection against infection. These findings illustrate that γδ T cells play a role with hallmarks of adaptive immunity in the intestinal mucosa.


Subject(s)
Immunologic Memory/immunology , Intestines/immunology , Receptors, Antigen, T-Cell, gamma-delta/immunology , T-Lymphocytes/immunology , Adaptive Immunity/immunology , Amino Acid Sequence , Animals , Cells, Cultured , Female , Flow Cytometry , Host-Pathogen Interactions/immunology , Hyaluronan Receptors/immunology , Hyaluronan Receptors/metabolism , Interferon-gamma/immunology , Interferon-gamma/metabolism , Interleukin-17/immunology , Interleukin-17/metabolism , Intestinal Mucosa/immunology , Intestinal Mucosa/metabolism , Listeria monocytogenes/immunology , Listeria monocytogenes/physiology , Listeriosis/immunology , Listeriosis/metabolism , Mice , Mice, Congenic , Mice, Inbred BALB C , Receptors, Antigen, T-Cell, gamma-delta/genetics , Receptors, Antigen, T-Cell, gamma-delta/metabolism , T-Lymphocyte Subsets/immunology , T-Lymphocyte Subsets/metabolism , T-Lymphocytes/metabolism , Tumor Necrosis Factor Receptor Superfamily, Member 7/immunology , Tumor Necrosis Factor Receptor Superfamily, Member 7/metabolism
5.
PLoS One ; 8(2): e56539, 2013.
Article in English | MEDLINE | ID: mdl-23409193

ABSTRACT

Programmed death ligand-1 (PD-L1) is an important negative regulator of T cell immune responses via interactions with PD-1 and CD80. However, PD-L1 can also act as a positive costimulator, but the relevant counterreceptor is not known. We analyzed the role of PD-L1 in CD8-T cell responses to infection with Listeria monocytogenes (LM) or vesicular stomatitis virus (VSV). PD-L1 blockade impaired antigen-specific CD8 effector T cell expansion in response to LM, but not to VSV infection, particularly limiting short-lived effector cell differentiation. Simultaneous CD4-T cell depletion and anti-PD-L1 blockade revealed that PD-L1 provided costimulation even in the absence of CD4-T cells. Most importantly, specific blockade of PD-L1 binding to CD80 or to PD-1 did not recapitulate PDL-1 blockade. The results suggested that PD-L1 plays an important costimulatory role for antigen-specific CD8 T cells during LM infection perhaps through a distinct receptor or interaction epitope.


Subject(s)
B7-H1 Antigen/immunology , CD8-Positive T-Lymphocytes/immunology , Listeria monocytogenes/immunology , Listeriosis/immunology , Signal Transduction , Animals , Antibodies, Monoclonal/immunology , Antigens, Differentiation/immunology , B7-1 Antigen/immunology , B7-H1 Antigen/metabolism , CD8-Positive T-Lymphocytes/cytology , Cell Differentiation , Cell Proliferation , Epitopes, T-Lymphocyte/immunology , Listeriosis/microbiology , Mice , Mice, Inbred C57BL , Programmed Cell Death 1 Receptor , Rhabdoviridae Infections/immunology , Rhabdoviridae Infections/virology , T-Lymphocyte Subsets/cytology , T-Lymphocyte Subsets/immunology , Up-Regulation , Vesiculovirus/immunology
6.
Am J Reprod Immunol ; 65(4): 438-47, 2011 Apr.
Article in English | MEDLINE | ID: mdl-20825380

ABSTRACT

PROBLEM: The NFκB pathway is a major source of pro-inflammatory cytokines, which may contribute to cancer chemoresistance. We showed that constitutive NFκB activity is characteristic of the ovarian cancer stem cells (OCSCs). The aim of this study is to determine whether the inhibition of NFκB by Eriocalyxin B (EriB) in the OCSCs may induce cell death in otherwise chemoresistant cells. METHODS: OCSCs and mature ovarian cancer cells (mOCCs) were treated with increasing concentrations of EriB. Cell viability was measured using the Celltiter 96 assay, and caspase activity was quantified using Caspase-Glo™ assay. Cytokine levels were quantified using xMAP technology. RESULTS: EriB decreased the percent of viable cells in all cultures tested with GI(50) of 0.5-1 µm after 48 hrs of treatment. The intracellular changes associated with EriB-induced cell death are: (i) inhibition of NF-κB activity; (ii) decreased cytokine production; (iii) activation of caspases; and (iv) down-regulation of XIAP. In addition, EriB is able to sensitize OCSCs to TNFα and FasL-mediated cell death. CONCLUSION: Inhibition of the NFκB pathway induces cell death in the OCSCs. Because the OCSCs may represent the source of recurrence and chemoresistance, the use of NFκB inhibitors like EriB may prevent recurrence in patients with ovarian cancer.


Subject(s)
Diterpenes/pharmacology , NF-kappa B/metabolism , Neoplastic Stem Cells/immunology , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/metabolism , Apoptosis/drug effects , Blotting, Western , Caspases/metabolism , Cell Survival/drug effects , Cells, Cultured , Cytokines/biosynthesis , Electrophoresis, Polyacrylamide Gel , Female , Humans , NF-kappa B/antagonists & inhibitors , Neoplastic Stem Cells/drug effects , Ovarian Neoplasms/immunology , Ovarian Neoplasms/pathology , Tumor Necrosis Factor-alpha/antagonists & inhibitors , X-Linked Inhibitor of Apoptosis Protein/genetics
7.
BMC Cancer ; 10: 349, 2010 Jul 02.
Article in English | MEDLINE | ID: mdl-20598131

ABSTRACT

BACKGROUND: Development of innovative, effective therapies against recurrent/chemotherapy-resistant ovarian cancer remains a high priority. Using high-throughput technologies to analyze genetic fingerprints of ovarian cancer, we have discovered extremely high expression of the genes encoding the proteins claudin-3 and claudin-4. METHODS: Because claudin-3 and -4 are the epithelial receptors for Clostridium perfringens enterotoxin (CPE), and are sufficient to mediate CPE binding, in this study we evaluated the in vitro and in vivo bioactivity of the carboxy-terminal fragment of CPE (i.e., CPE290-319 binding peptide) as a carrier for tumor imaging agents and intracellular delivery of therapeutic drugs. Claudin-3 and -4 expression was examined with rt-PCR and flow cytometry in multiple primary ovarian carcinoma cell lines. Cell binding assays were used to assess the accuracy and specificity of the CPE peptide in vitro against primary chemotherapy-resistant ovarian carcinoma cell lines. Confocal microscopy and biodistribution assays were performed to evaluate the localization and uptake of the FITC-conjugated CPE peptide in established tumor tissue. RESULTS: Using a FITC-conjugated CPE peptide we show specific in vitro and in vivo binding to multiple primary chemotherapy resistant ovarian cancer cell lines. Bio-distribution studies in SCID mice harboring clinically relevant animal models of chemotherapy resistant ovarian carcinoma showed higher uptake of the peptide in tumor cells than in normal organs. Imunofluorescence was detectable within discrete accumulations (i.e., tumor spheroids) or even single chemotherapy resistant ovarian cancer cells floating in the ascites of xenografted animals while a time-dependent internalization of the FITC-conjugated CPE peptide was consistently noted in chemotherapy-resistant ovarian tumor cells by confocal microscopy. CONCLUSIONS: Based on the high levels of claudin-3 and -4 expression in chemotherapy-resistant ovarian cancer and other highly aggressive human epithelial tumors including breast, prostate and pancreatic cancers, CPE peptide holds promise as a lead peptide for the development of new diagnostic tracers or alternative anticancer agents.


Subject(s)
Adenocarcinoma, Clear Cell/drug therapy , Adenocarcinoma/drug therapy , Carcinoma, Papillary/drug therapy , Carcinoma, Squamous Cell/drug therapy , Cystadenocarcinoma, Serous/drug therapy , Enterotoxins/pharmacology , Ovarian Neoplasms/drug therapy , Peptide Fragments/pharmacology , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma, Clear Cell/genetics , Adenocarcinoma, Clear Cell/metabolism , Animals , Carcinoma, Papillary/genetics , Carcinoma, Papillary/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Chlorocebus aethiops , Claudin-3 , Claudin-4 , Clostridium perfringens , Cystadenocarcinoma, Serous/genetics , Cystadenocarcinoma, Serous/metabolism , Drug Resistance, Neoplasm , Female , Fibroblasts , Flow Cytometry , Humans , Membrane Proteins/genetics , Membrane Proteins/metabolism , Mice , Mice, SCID , Ovarian Neoplasms/genetics , Ovarian Neoplasms/metabolism , Peptide Fragments/pharmacokinetics , RNA, Messenger/genetics , Reverse Transcriptase Polymerase Chain Reaction , Spheroids, Cellular/drug effects , Tissue Distribution , Transplantation, Heterologous , Tumor Cells, Cultured , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics , Uterine Cervical Neoplasms/metabolism , Vero Cells
8.
Stem Cells ; 27(10): 2405-13, 2009 Oct.
Article in English | MEDLINE | ID: mdl-19658191

ABSTRACT

Neovascularization is required for solid tumor maintenance, progression, and metastasis. The most described contribution of cancer cells in tumor neovascularization is the secretion of factors, which attract various cell types to establish a microenvironment that promotes blood vessel formation. The cancer stem cell hypothesis suggests that tumors are composed of cells that may share the differentiation capacity of normal stem cells. Similar to normal stem cells, cancer stem cells (CSCs) have the capacity to acquire different phenotypes. Thus, it is possible that CSCs have a bigger role in the process of tumor neovascularization. In this study, we show the capacity of a specific population of ovarian cancer cells with stem-like properties to give rise to xenograft tumors containing blood vessels, which are lined by human CD34+ cells. In addition, when cultured in high-density Matrigel, these cells mimic the behavior of normal endothelial cells and can form vessel-like structures in 24 hours. Microscopic analysis showed extensive branching and maturation of vessel-like structures in 7 days. Western blot and flow cytometry analysis showed that this process is accompanied by the acquisition of classic endothelial markers, CD34 and VE-cadherin. More importantly, we show that this process is vascular endothelial growth factor-independent, but IKK beta-dependent. Our findings suggest that anti-angiogenic therapies should take into consideration the inherent capacity of these cells to serve as vascular progenitors.


Subject(s)
Carcinoma/blood supply , Carcinoma/physiopathology , Mesenchymal Stem Cells/physiology , Neoplastic Stem Cells/physiology , Neovascularization, Pathologic/physiopathology , Ovarian Neoplasms/blood supply , Ovarian Neoplasms/physiopathology , Animals , Antigens, CD/metabolism , Antigens, CD34/metabolism , Biomarkers/metabolism , Blood Vessels/cytology , Blood Vessels/metabolism , Cadherins/metabolism , Carcinoma/metabolism , Endothelial Cells/cytology , Endothelial Cells/metabolism , Female , Humans , I-kappa B Kinase/metabolism , Mice , Mice, Nude , Ovarian Neoplasms/metabolism , Transplantation, Heterologous , Tumor Cells, Cultured
9.
Cancer ; 115(14): 3204-16, 2009 Jul 15.
Article in English | MEDLINE | ID: mdl-19472400

ABSTRACT

BACKGROUND: Resistance to apoptosis is 1 of the key events that confer chemoresistance and is mediated by the overexpression of antiapoptotic proteins, which inhibit caspase activation. The objective of this study was to evaluate whether the activation of an alternative, caspase-independent cell death pathway could promote death in chemoresistant ovarian cancer cells. The authors report the characterization of NV-128 as an inducer of cell death through a caspase-independent pathway. METHODS: Primary cultures of epithelial ovarian cancer (EOC) cells were treated with increasing concentration of NV-128, and the concentration that caused 50% growth inhibition (GI(50)) was determined using a proprietary assay. Apoptotic proteins were characterized by Western blot analyses, assays that measured caspase activity, immunohistochemistry, and flow cytometry. Protein-protein interactions were determined using immunoprecipitation. In vivo activity was measured in a xenograft mice model. RESULTS: NV-128 was able to induce significant cell death in both paclitaxel-resistant and carboplatin-resistant EOC cells with a GI(50) between 1 microg/mL and 5 microg/mL. Cell death was characterized by chromatin condensation but was caspase-independent. The activated pathway involved the down-regulation of phosphorylated AKT, phosphorylated mammalian target of rapamycin (mTOR), and phosphorylated ribosomal p70 S6 kinase, and the mitochondrial translocation of beclin-1 followed by nuclear translocation of endonuclease G. CONCLUSIONS: The authors characterized a novel compound, NV-128, which inhibits mTOR and promotes caspase-independent cell death. The current results indicated that inhibition of mTOR may represent a relevant pathway for the induction of cell death in cells resistant to the classic caspase-dependent apoptosis. These findings demonstrate the possibility of using therapeutic drugs, such as NV-128, which may have beneficial effects in patients with chemoresistant ovarian cancer.


Subject(s)
Antineoplastic Agents/pharmacology , Isoflavones/pharmacology , Oncogene Protein v-akt/metabolism , Ovarian Neoplasms/pathology , Protein Kinases/metabolism , Signal Transduction , Animals , Antineoplastic Agents/toxicity , Autophagy , Caspases/pharmacology , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Chromatin/drug effects , Down-Regulation , Female , Humans , Isoflavones/toxicity , Mice , Mitochondria/drug effects , Ovarian Neoplasms/metabolism , Phosphorylation , TOR Serine-Threonine Kinases , Xenograft Model Antitumor Assays
10.
Cell Cycle ; 8(1): 158-66, 2009 Jan 01.
Article in English | MEDLINE | ID: mdl-19158483

ABSTRACT

A major burden in the treatment of ovarian cancer is the high percentage of recurrence and chemoresistance. Cancer stem cells (CSCs) provide a reservoir of cells that can self-renew, can maintain the tumor by generating differentiated cells [non-stem cells (non-CSCs)] which make up the bulk of the tumor and may be the primary source of recurrence. We describe the characterization of human ovarian cancer stem cells (OCSCs). These cells have a distinctive genetic profile that confers them with the capacity to recapitulate the original tumor, proliferate with chemotherapy, and promote recurrence. CSC identified in EOC cells isolated form ascites and solid tumors are characterized by: CD44+, MyD88+, constitutive NFkappaB activity and cytokine and chemokine production, high capacity for repair, chemoresistance to conventional chemotherapies, resistance to TNFalpha-mediated apoptosis, capacity to form spheroids in suspension, and the ability to recapitulate in vivo the original tumor. Chemotherapy eliminates the bulk of the tumor but it leaves a core of cancer cells with high capacity for repair and renewal. The molecular properties identified in these cells may explain some of the unique characteristics of CSCs that control self-renewal and drive metastasis. The identification and cloning of human OCSCs can aid in the development of better therapeutic approaches for ovarian cancer patients.


Subject(s)
Drug Resistance, Neoplasm , Neoplastic Stem Cells/pathology , Ovarian Neoplasms/pathology , Ovarian Neoplasms/therapy , Animals , Cell Differentiation , Cell Proliferation , Cytokines/biosynthesis , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Hyaluronan Receptors/metabolism , Mice , Myeloid Differentiation Factor 88/metabolism , NF-kappa B/metabolism , Neoplastic Stem Cells/metabolism , Ovarian Neoplasms/genetics , Phenotype , Spheroids, Cellular/metabolism , Spheroids, Cellular/pathology , Survival Analysis , Toll-Like Receptor 4/metabolism , Tumor Cells, Cultured
11.
Ann N Y Acad Sci ; 1127: 129-33, 2008 Apr.
Article in English | MEDLINE | ID: mdl-18443340

ABSTRACT

Preeclampsia is associated with an increased release of factors from the placental syncytium into maternal blood, including the antiangiogenic factors soluble fms-like tyrosine kinase-1 and soluble endoglin, the antifibrinolytic factor plasminogen activator inhibitor-1, prostanoids, lipoperoxides, cytokines, and microparticles. These factors are suggested to promote maternal endothelium dysfunction and are associated with placental damage in pregnancies also complicated with intrauterine growth restriction (IUGR). In this report, we briefly describe the interaction of syncytial factors with hypoxia, reactive oxygen species, and apoptosis in the pathophysiology of preeclampsia and IUGR. Given the critical role of the syncytium in these complications of pregnancy, we also present a novel methodology in which laser capture microdissection followed by Western blotting is used to assess levels of syncytial Fas ligand, a key protein in the apoptotic cascade.


Subject(s)
Fetal Growth Retardation/pathology , Gene Expression Regulation, Developmental , Giant Cells/metabolism , Neovascularization, Physiologic , Placenta/metabolism , Pre-Eclampsia/pathology , Apoptosis , Fas Ligand Protein , Female , Fetal Growth Retardation/diagnosis , Humans , Lasers , Models, Biological , Pre-Eclampsia/diagnosis , Pregnancy , Reactive Oxygen Species , Trophoblasts/metabolism
12.
Methods Mol Biol ; 414: 35-45, 2008.
Article in English | MEDLINE | ID: mdl-18175810

ABSTRACT

Tumors are heterogeneous structures that contain different cell populations. Laser capture microdissection (LCM) can be used to obtain pure cancer cells from fresh-frozen cancer tissue and the surrounded environment, thus providing an accurate snapshot of the tumor and its microenvironment in vivo. We describe a new approach to isolate pure cancer cell population and evaluate protein expression. The process includes immunocytochemistry, laser microdissection, and western blot analysis. Using this technique, we can detect proteins such as X-linked inhibitor of apoptosis protein (XIAP) and Fas ligand (FasL) with as little as 1000 cells.


Subject(s)
Cryopreservation , Lasers , Microdissection/methods , Neoplasm Proteins/analysis , Ovarian Neoplasms/pathology , Biomarkers, Tumor/analysis , Cryoultramicrotomy/methods , Fas Ligand Protein/analysis , Female , Humans , Immunohistochemistry/methods , Myeloid Differentiation Factor 88/analysis , X-Linked Inhibitor of Apoptosis Protein/analysis
13.
Food Chem Toxicol ; 44(9): 1513-21, 2006 Sep.
Article in English | MEDLINE | ID: mdl-16765500

ABSTRACT

Bidens pilosa (B. pilosa) is well known in Taiwan as a traditional Chinese medicine. The purpose of this study was to evaluate the ability of both the ethanol (EtOH) and ethylacetate/ethanol (EA/EtOH) extracts from the whole B. pilosa plant, to protect normal human erythrocytes against oxidative damage in vitro. It was determined that the oxidative hemolysis and lipid/protein peroxidation of erythrocytes induced by the aqueous peroxyl radical [2,2'-azobis(2-amidinopropane) dihydrochloride (AAPH)] were suppressed by both EtOH (50-150 microg/ml) and EA/EtOH (25-75 microg/ml) extracts of B. pilosa in concentration- and time-dependent manners. B. pilosa extracts also prevented the decline of superoxide dismutase (SOD) activity and the depletion of cytosolic glutathione (GSH) and ATP in erythrocytes. These results imply that B. pilosa may have protective antioxidant properties.


Subject(s)
Antioxidants/pharmacology , Bidens , Drugs, Chinese Herbal/pharmacology , Erythrocytes/drug effects , Oxidative Stress/drug effects , Acetates/chemistry , Adenosine Triphosphate/metabolism , Bidens/chemistry , Cells, Cultured , Cytosol/drug effects , Cytosol/metabolism , Dose-Response Relationship, Drug , Electrophoresis, Polyacrylamide Gel , Erythrocyte Membrane/drug effects , Erythrocyte Membrane/metabolism , Erythrocytes/metabolism , Ethanol/chemistry , Glutathione/metabolism , Humans , Membrane Proteins/chemistry , Superoxide Dismutase/metabolism
14.
Curr Opin Investig Drugs ; 7(6): 542-8, 2006 Jun.
Article in English | MEDLINE | ID: mdl-16784025

ABSTRACT

Epithelial ovarian cancer is the fourth leading cause of cancer-related deaths in women and is the most lethal of the gynecological malignancies. Thle high mortality rate arises from difficulties in the early detection of the disease and the widespread development of chemoresistance. Phenoxodiol, a novel isoflavone derivative, has demonstrated antitumor activity. In addition, it has been shown to induce cell death in chemoresistant epithelial ovarian cancer cells. Moreover, suboptimal exposure of these cells to phenoxodiol lowered the IC50 value of numerous chemotherapeutic agents. In this review, the current understanding of the mechanism of action of phenoxodiol, its potential clinical application for the treatment of ovarian cancer and the concept of chemosensitization are discussed.


Subject(s)
Antineoplastic Agents/therapeutic use , Isoflavones/therapeutic use , Ovarian Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Caspase 3 , Caspases/metabolism , Cells, Cultured , Clinical Trials as Topic , Death Domain Receptor Signaling Adaptor Proteins , Drug Evaluation, Preclinical , Drug Resistance, Neoplasm , Female , Humans , Isoflavones/pharmacology , Ovarian Neoplasms/metabolism , Ovarian Neoplasms/pathology , Randomized Controlled Trials as Topic , Tumor Necrosis Factor Receptor-Associated Peptides and Proteins/metabolism
15.
Yale J Biol Med ; 79(3-4): 153-63, 2006 Dec.
Article in English | MEDLINE | ID: mdl-17940625

ABSTRACT

INTRODUCTION: Early identification of chemoresistance in patients with ovarian cancer is of utmost importance in order to provide them with the most appropriate therapy. Recently, we described the expression of MyD88 in ovarian cancer cells that were resistant to the cytotoxic agent paclitaxel. In addition to chemoresistance, in MyD88 positive ovarian cancer cells, paclitaxel stimulates growth and production of proinflammatory cytokines. The objective of this study was to determine the correlation of MyD88 expression in primary and recurrent epithelial ovarian cancers with the response to carboplatin and paclitaxel combination chemotherapy. METHODS: Tumors are heterogeneous structures that contain different cell populations, thus rendering the identification of specific tumor markers difficult. Using laser capture microdissection, pure cancer cells were isolated from ovarian malignant tumors that were obtained from 20 patients at the time of surgery. The microdissected cells were evaluated for the expression of MyD88, FasL, and XIAP by western blot analysis. RESULTS: Protein expression was observed in samples containing as low as 500 cells. The results were correlated with the clinical course of those patients. It was evident that MyD88 expression in ovarian cancer cells accurately predicts a poor response to paclitaxel chemotherapy as shown by a short progression-free interval and overall survival. CONCLUSION: We describe for the first time a molecular approach to identify paclitaxel chemoresistance. Toxicity from agents without therapeutic benefit can be avoided by identifying those patients who will not respond to a specific agent. Molecular markers will enable us to design individualized treatments and improve overall survival.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Drug Resistance, Neoplasm , Gene Expression Regulation, Neoplastic , Myeloid Differentiation Factor 88/biosynthesis , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/metabolism , Paclitaxel/pharmacology , Disease-Free Survival , Female , Humans , Lasers , Microdissection , Treatment Outcome , Tumor Cells, Cultured
SELECTION OF CITATIONS
SEARCH DETAIL
...