Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 44
Filter
1.
Front Endocrinol (Lausanne) ; 14: 1223312, 2023.
Article in English | MEDLINE | ID: mdl-37492197

ABSTRACT

Introduction: We successfully developed a broad spectrum of patient-derived endocrine organoids (PDO) from benign and malignant neoplasms of thyroid, parathyroid, and adrenal glands. In this study, we employed functionally intact parathyroid PDOs from benign parathyroid tissues to study primary hyperparathyroidism (PHPT), a common endocrine metabolic disease. As proof of concept, we examined the utility of parathyroid PDOs for bioenergetic and metabolic screening and assessed whether parathyroid PDO metabolism recapitulated matched PHPT tissues. Methods: Our study methods included a fine-needle aspiration (FNA)-based technique to establish parathyroid PDOs from human PHPT tissues (n=6) in semi-solid culture conditions for organoid formation, growth, and proliferation. Mass spectrometry metabolomic analysis of PHPT tissues and patient-matched PDOs, and live cell bioenergetic profiling of parathyroid PDOs with extracellular flux analyses, were performed. Functional analysis cryopreserved and re-cultured parathyroid PDOs for parathyroid hormone (PTH) secretion was performed using ELISA hormone assays. Results and discussion: Our findings support both the feasibility of parathyroid PDOs for metabolic and bioenergetic profiling and reinforce metabolic recapitulation of PHPT tissues by patient-matched parathyroid PDOs. Cryopreserved parathyroid PDOs exhibited preserved, rapid, and sustained secretory function after thawing. In conclusion, successful utilization of parathyroid PDOs for metabolic profiling further affirms the feasibility of promising endocrine organoid platforms for future metabolic studies and broader multiplatform and translational applications for therapeutic advancements of parathyroid and other endocrine applications.


Subject(s)
Parathyroid Glands , Thyroid Gland , Humans , Parathyroid Glands/metabolism , Biopsy, Fine-Needle/methods , Organoids
2.
Cancer Res ; 83(10): 1573-1580, 2023 05 15.
Article in English | MEDLINE | ID: mdl-36877156

ABSTRACT

Nucleophosmin (NPM1) is frequently mutated in acute myeloid leukemia, and NPM1 expression is elevated in several cancer types. NPM1 is a multifunctional oligomeric protein involved in numerous cellular functions that include participating in liquid-liquid phase separation, ribosome biogenesis, chaperoning of histones, and modulation of transcription. In this review, we discuss the underappreciated role of NPM1 in DNA damage repair, specifically Polη-mediated translesion synthesis, base excision, and homologous recombination and highlight the therapeutic potential of NPM1 targeting in cancer treatment.


Subject(s)
Leukemia, Myeloid, Acute , Nucleophosmin , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Mutation , DNA Damage , DNA Repair , Leukemia, Myeloid, Acute/therapy , Leukemia, Myeloid, Acute/drug therapy
3.
Surgery ; 173(1): 67-75, 2023 Jan.
Article in English | MEDLINE | ID: mdl-36400581

ABSTRACT

BACKGROUND: Recent advancements in 3-dimensional patient-derived organoid models have revolutionized the field of cancer biology. There is an urgent need for development of endocrine tumor organoid models for medullary thyroid carcinoma, adrenocortical carcinoma, papillary thyroid carcinoma, and a spectrum of benign hyperfunctioning parathyroid and adrenal neoplasms. We aimed to engineer functionally intact 3-dimensional endocrine patient-derived organoids to expand the in vitro and translational applications for the advancement of endocrine research. METHODS: Using our recently developed fine needle aspiration-based methodology, we established patient-derived 3-dimensional endocrine organoid models using prospectively collected human papillary thyroid carcinoma (n = 6), medullary thyroid carcinoma (n = 3), adrenocortical carcinoma (n = 3), and parathyroid (n = 5). and adrenal (n = 5) neoplasms. Multiplatform analyses of endocrine patient-derived organoids and applications in oncoimmunology, near-infrared autofluorescence, and radiosensitization studies under 3-dimensional in vitro conditions were performed. RESULTS: We have successfully modeled and analyzed the complex endocrine microenvironment for a spectrum of endocrine neoplasms in 3-dimensional culture. The endocrine patient-derived organoids recapitulated complex tumor microenvironment of endocrine neoplasms morphologically and functionally and maintained cytokine production and near-infrared autofluorescence properties. CONCLUSION: Our novel engineered endocrine patient-derived organoid models of thyroid, parathyroid and adrenal neoplasms represent an exciting and elegant alternative to current limited 2-dimensional systems and afford future broad multiplatform in vitro and translational applications, including in endocrine oncoimmunology.


Subject(s)
Adrenal Gland Neoplasms , Thyroid Neoplasms , Humans , Organoids , Tumor Microenvironment , Thyroid Neoplasms/pathology , Adrenal Gland Neoplasms/pathology
4.
World J Surg ; 47(2): 371-381, 2023 02.
Article in English | MEDLINE | ID: mdl-36195678

ABSTRACT

PURPOSE: Papillary thyroid carcinoma (PTC) progression imparts reduced patient survival. Tumor resistance and progression can be influenced by Glutathione (GSH) metabolism. Glutathione peroxidase 4 (GPX4) regulates GSH oxidation to prevent lipid peroxidation of cell membranes during increased oxidative stress and regulates ferroptosis cell death pathway in tumor cells. This study examines the differential ferroptosis effects by GPX4 inhibitors in thyroid cancer cell and 3-D spheroid in vitro models. MATERIALS AND METHODS: We examined differential effects of GPX4 inhibitors on PTC cells (K1, MDA-T32, MDA-T68) with BRAF and RAS mutations, and TERT promoter and PIK3CA co-mutations. The effects of GPX4 inhibitors on ferroptosis activation, proliferation, oxidative stress, and activation of signaling pathways were assessed by Western blot, total (GSH) and oxidized glutathione (GSSG) levels, ROS induction, RT-qPCR, migration, and proliferation assays. RESULTS: GPX4 inhibitors induced ferroptosis, rising ROS, GSH depletion, arrested tumor cell migration, increased DNA damage, suppressed mTOR pathway and DNA repair response in PTC cells in vitro. Differential responses to DNA damage and GPX4 levels were observed between 3-D PTC spheroids and thyroid cancer cells in a monolayer model. CONCLUSION: Effective GPX4 inhibition with various inhibitors induced a robust but differential activation of ferroptosis in monolayer thyroid tumor cell and 3-D PTC spheroid models. Our study is the first of its kind to determine the differential effects of GPX4 inhibitors on thyroid cancer cells with diverse mutational signatures. We have identified a novel mechanism of action of GPX4 inhibition in preclinical in vitro models of thyroid cancer that can be further exploited for therapeutic benefit in advanced therapy-resistant thyroid cancers.


Subject(s)
Ferroptosis , Thyroid Neoplasms , Humans , Phospholipid Hydroperoxide Glutathione Peroxidase/metabolism , Glutathione Peroxidase/genetics , Glutathione Peroxidase/metabolism , Reactive Oxygen Species/metabolism , Thyroid Neoplasms/drug therapy , Thyroid Neoplasms/genetics
5.
Sci Rep ; 12(1): 19396, 2022 11 12.
Article in English | MEDLINE | ID: mdl-36371529

ABSTRACT

Papillary thyroid carcinoma (PTC) demonstrates significantly reduced patient survival with metastatic progression. Tumor progression can be influenced by metabolism, including antioxidant glutathione (GSH). Glutathione peroxidase 4 (GPX4) is a selenoenzyme that uses GSH as a co-factor to regulate lipid peroxidation of cell membranes during increased oxidative stress. GPX4 suppression in tumor cells can induce ferroptosis. This study aims to examine ferroptosis as a potentially critical pathway in effective targeting of thyroid cancer (TC) cells. We treated human TC cells (K1, MDA-T68, MDA-T32, TPC1) with (1S,3R)-RSL3 (RSL3), a small-molecule inhibitor of GPX4 and examined the effects on ferroptosis, tumor cell survival and migration, spheroid formation, oxidative stress, DNA damage repair response, and mTOR signaling pathway in vitro. GPX4 inhibition activated ferroptosis, inducing TC cell death, rapid rise in reactive oxygen species and effectively arrested cell migration in vitro. Suppression of mTOR signaling pathway triggered autophagy. GPX4 genetic knockdown mirrored RSL3 effect on mTOR pathway suppression. RSL3 subdued DNA damage repair response by suppressing phosphorylation of nucleophosmin 1 (NPM1). Thus, observed potent induction of ferroptosis, GPX4-dependent novel suppression of mTOR pathway and DNA damage repair response in preclinical in vitro model of TC supports GPX4 targeting for therapeutic benefit in advanced therapy-resistant thyroid cancers.


Subject(s)
Ferroptosis , Thyroid Neoplasms , Humans , Phospholipid Hydroperoxide Glutathione Peroxidase , Cell Death , Glutathione Peroxidase/metabolism , Glutathione/metabolism , Thyroid Neoplasms/drug therapy , TOR Serine-Threonine Kinases
6.
Surgery ; 171(1): 111-118, 2022 01.
Article in English | MEDLINE | ID: mdl-34261605

ABSTRACT

BACKGROUND: Immunotherapeutic response failure of adrenocortical carcinomas highlights a need for novel strategies targeting immune cell populations in the tumor microenvironment to overcome tumor resistance and enhance therapeutic response. A recent study explored a new link between tumor mast cell infiltration and improved outcomes in patients with adrenocortical carcinomas. We further dissect the role of mast cells in the tumor microenvironment of adrenocortical carcinomas by examining the tumor mast cell expression signatures and mast cell activity within the tumor microenvironment to provide additional insight into potential novel immunotherapeutic targets. METHODS: Using the CIBERSORTx computational immunogenomic deconvolution algorithm to analyze adrenocortical carcinoma tumor gene messenger RNA expression data (The Cancer Genome Atlas, N = 79), we estimated the abundance of tumor immune infiltrating mast cells and assessed prognostic potential of mast cell signaling genes as pro or antitumor signatures, as well as examined the impact on overall and disease-free survival. RESULTS: We stratified mast cell signaling genes with survival prognostic values (overall survival, disease-free survival, P < .05) into antitumor (ALOX5, CCL2, CCL5, CXCL10, HDC, IL16, TNF, TPSAB1, VEGFD) and protumor (CXCL1, CXCL3, CXCL8, IL4, IL13, PTGS3, TNSF4, VEGFD) groups. Antitumor mast cell signature, as the predominant phenotype, was associated with improved overall and disease-free survival. CONCLUSION: The deconvolution analysis of The Cancer Genome Atlas data identified mast cell infiltration in the adrenocortical carcinoma microenvironment as predominantly associated with antitumor activity. Future studies stemming from our findings may help define the role of mast cells in the tumor microenvironment and the impact on patient survival in patients with adrenocortical carcinomas. Modulation of tumor mast cell infiltration may serve as a potential target for novel synergistic immunotherapies for the treatment and improved survival of patients with adrenocortical carcinomas.


Subject(s)
Adrenal Cortex Neoplasms/genetics , Adrenocortical Carcinoma/genetics , Gene Expression Regulation, Neoplastic/immunology , Mast Cells/immunology , Neoplasm Recurrence, Local/epidemiology , Adrenal Cortex/immunology , Adrenal Cortex/pathology , Adrenal Cortex Neoplasms/immunology , Adrenal Cortex Neoplasms/mortality , Adrenal Cortex Neoplasms/therapy , Adrenalectomy , Adrenocortical Carcinoma/immunology , Adrenocortical Carcinoma/mortality , Adrenocortical Carcinoma/therapy , Adult , Aged , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Chemotherapy, Adjuvant/methods , Disease-Free Survival , Drug Synergism , Female , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Mast Cells/metabolism , Middle Aged , Neoadjuvant Therapy/methods , Prognosis , Retrospective Studies , Tumor Microenvironment/drug effects , Tumor Microenvironment/genetics , Tumor Microenvironment/immunology
7.
J Cell Mol Med ; 25(21): 10061-10072, 2021 11.
Article in English | MEDLINE | ID: mdl-34664400

ABSTRACT

Adrenocortical carcinoma (ACC) is a rare but highly aggressive malignancy. Nearly half of ACC tumours overproduce and secrete adrenal steroids. Excess cortisol secretion, in particular, has been associated with poor prognosis among ACC patients. Furthermore, recent immunotherapy clinical trials have demonstrated significant immunoresistance among cortisol-secreting ACC (CS-ACC) patients when compared to their non-cortisol-secreting (nonCS-ACC) counterparts. The immunosuppressive role of excess glucocorticoid therapies and hypersecretion is known; however, the impact of the cortisol hypersecretion on ACC tumour microenvironment (TME), immune expression profiles and immune cell responses remain largely undefined. In this study, we characterized the TME of ACC patients and compared the immunogenomic profiles of nonCS-ACC and CS-ACC tumours to assess the impact of differentially expressed genes (DEGs) by utilizing The Cancer Genome Atlas (TCGA) database. Immunogenomic comparison (CS- vs. nonCS-ACC tumour TMEs) demonstrated an immunosuppressive expression profile with a direct impact on patient survival. We identified several primary prognostic indicators and potential targets within ACC tumour immune landscape. Differentially expressed immune genes with prognostic significance provide additional insight into the understanding of potential contributory mechanisms underlying failure of initial immunotherapeutic trials and poor prognosis of patients with CS-ACC.


Subject(s)
Adrenal Cortex Neoplasms/etiology , Adrenal Cortex Neoplasms/metabolism , Adrenocortical Carcinoma/etiology , Adrenocortical Carcinoma/metabolism , Computational Biology , Hydrocortisone/metabolism , Adrenal Cortex Neoplasms/pathology , Adrenocortical Carcinoma/pathology , Computational Biology/methods , Databases, Genetic , Disease Susceptibility , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Lymphocytes, Tumor-Infiltrating/immunology , Lymphocytes, Tumor-Infiltrating/metabolism , Lymphocytes, Tumor-Infiltrating/pathology , Tumor-Associated Macrophages/immunology , Tumor-Associated Macrophages/metabolism , Tumor-Associated Macrophages/pathology
8.
Cancer Lett ; 500: 220-227, 2021 03 01.
Article in English | MEDLINE | ID: mdl-33358698

ABSTRACT

The ability of chemo-radiation therapy to control locally advanced stage III non-small cell lung cancer (NSCLC) is poor. While addition of consolidation immunotherapy has improved outcomes in subsets of patients there is still an urgent need for new therapeutic targets. Emerging research indicates that nucleophosmin1 (NPM1) is over-expressed in NSCLC, promotes tumor growth and that over-expression correlates with a lower survival probability. NPM1 is critical for APE1 base excision activity and for RAD51-mediated repair of DNA double strand breaks (DSBs). YTR107 is a small molecule radiation sensitizer that has been shown to bind to NPM1, suppressing pentamer formation. Here we show that in irradiated cells YTR107 inhibits SUMOylated NPM1 from associating with RAD51, RAD51 foci formation and repair of DSBs. YTR107 acts synergistically with the PARP1/2 inhibitor ABT 888 to increase replication stress and radiation-induced cell lethality. YTR107 was found to radiosensitize tumor initiating cells. Congruent with this knowledge, adding YTR107 to a fractionated irradiation regimen diminished NSCLC xenograft growth and increased overall survival. These data support the hypothesis that YTR107 represents a therapeutic target for control of NSCLC.


Subject(s)
Carcinoma, Non-Small-Cell Lung/drug therapy , Carcinoma, Non-Small-Cell Lung/radiotherapy , DNA-(Apurinic or Apyrimidinic Site) Lyase/genetics , Nuclear Proteins/genetics , Rad51 Recombinase/genetics , Barbiturates/pharmacology , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Breaks, Double-Stranded/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , Humans , Indoles/pharmacology , Neoplasm Recurrence, Local/drug therapy , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/pathology , Neoplasm Recurrence, Local/radiotherapy , Nucleophosmin , Poly (ADP-Ribose) Polymerase-1/antagonists & inhibitors , Poly (ADP-Ribose) Polymerase-1/genetics , Radiation Tolerance/drug effects , Radiation-Sensitizing Agents/pharmacology , Sumoylation/drug effects , Sumoylation/radiation effects
9.
PLoS One ; 14(4): e0214670, 2019.
Article in English | MEDLINE | ID: mdl-30933998

ABSTRACT

Radiation therapy is often combined with androgen deprivation therapy in the treatment of aggressive localized prostate cancer. However, castration-resistant disease may not respond to testosterone deprivation approaches. Enzalutamide is a second-generation anti-androgen with high affinity and activity that is used for the treatment of metastatic disease. Although radiosensitization mechanisms are known to be mediated through androgen receptor activity, this project aims to uncover the detailed DNA damage repair factors influenced by enzalutamide using multiple models of androgen-sensitive (LNCaP) and castration-resistant human prostate cancer (22Rv1 and DU145). Enzalutamide is able to radiosensitize both androgen-dependent and androgen-independent human prostate cancer models in cell culture and xenografts in mice, as well as a treatment-resistant patient-derived xenograft. The enzalutamide-mediated mechanism of radiosensitization includes delay of DNA repair through temporal prolongation of the repair factor complexes and halting the cell cycle, which results in decreased colony survival. Altogether, these findings support the use of enzalutamide concurrently with radiotherapy to enhance the treatment efficacy for prostate cancer.


Subject(s)
DNA Repair/drug effects , Phenylthiohydantoin/analogs & derivatives , Prostatic Neoplasms , Radiation Tolerance , Radiation-Sensitizing Agents/pharmacology , Aged , Animals , Benzamides , Cell Line, Tumor , Cell Proliferation/drug effects , DNA Damage/drug effects , DNA Damage/genetics , DNA Repair/genetics , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Drug Resistance, Neoplasm/radiation effects , Humans , Male , Mice , Mice, Nude , Mice, Transgenic , Nitriles , Phenylthiohydantoin/pharmacology , Phenylthiohydantoin/therapeutic use , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Prostatic Neoplasms/radiotherapy , Prostatic Neoplasms, Castration-Resistant/drug therapy , Prostatic Neoplasms, Castration-Resistant/genetics , Prostatic Neoplasms, Castration-Resistant/pathology , Prostatic Neoplasms, Castration-Resistant/radiotherapy , Radiation Tolerance/drug effects , Radiation Tolerance/genetics , Signal Transduction/drug effects , Signal Transduction/genetics , Xenograft Model Antitumor Assays
10.
Radiat Res ; 190(2): 99-106, 2018 08.
Article in English | MEDLINE | ID: mdl-29799319

ABSTRACT

The transcription factor Nrf2 is an important modulator of antioxidant and drug metabolism, carbohydrate and lipid metabolism, as well as heme and iron metabolism. Regulation of Nrf2 expression occurs transcriptionally and post-transcriptionally. Post-transcriptional regulation entails ubiquitination followed by proteasome-dependent degradation. Additionally, Nrf2-mediated gene expression is subject to negative regulation by ATF3, Bach1 and cMyc. Nrf2-mediated gene expression is an important regulator of a cell's response to radiation. Although a majority of studies have shown that Nrf2 deficient cells are radiosensitized and Nrf2 over expression confers radioresistance, Nrf2's role in mediating the radiation response of crypt cells is controversial. The Nrf2 activator CDDO attenuates radiation-mediated crypt injury, whereas intestinal crypts in Nrf2 null mice are radiation resistant. Further investigation is needed in order to define the relationship between Nrf2 and radiation sensitivity in Lgr5+ and Bmi1+ cells that regulate regeneration of crypt stem cells. In hematopoietic compartments Nrf2 promotes the survival of irradiated osteoblasts that support long-term hematopoietic stem cell (LT-HSC) niches. Loss of Nrf2 in LT-HSCs increases stem cell intrinsic radiosensitivity, with the consequence of lowering the LD5030. An Nrf2 deficiency drives LT-HSCs from a quiescent to a proliferative state. This results in hematopoietic exhaustion and reduced engraftment after myoablative irradiation. The question of whether induction of Nrf2 in LT-HSC enhances hematopoietic reconstitution after bone marrow transplantation is not yet resolved. Irradiation of the lung induces pulmonary pneumonitis and fibrosis. Loss of Nrf2 promotes TGF-ß/Smad signaling that induces ATF3 suppression of Nrf2-mediated target gene expression. This, in turn, results in elevated reactive oxygen species (ROS) and isolevuglandin adduction of protein that impairs collagen degradation, and may contribute to radiation-induced chronic cell injury. Loss of Nrf2 impairs ΔNp63 stem/progenitor cell mobilization after irradiation, while promoting alveolar type 2 cell epithelial-mesenchymal transitions into myofibroblasts. These studies identify Nrf2 as an important factor in the radiation response of normal tissue.


Subject(s)
NF-E2-Related Factor 2/metabolism , Radiation Injuries/metabolism , Animals , Hematopoietic System/radiation effects , Humans , Intestinal Mucosa/metabolism , Intestines/radiation effects , Lung/metabolism , Lung/radiation effects , Radiation Injuries/pathology
11.
Free Radic Biol Med ; 112: 578-586, 2017 11.
Article in English | MEDLINE | ID: mdl-28870520

ABSTRACT

The development of radiation-induced pulmonary fibrosis represents a critical clinical issue limiting delivery of therapeutic doses of radiation to non-small cell lung cancer. Identification of the cell types whose injury initiates a fibrotic response and the underlying biological factors that govern that response are needed for developing strategies that prevent or mitigate fibrosis. C57BL/6 mice (wild type, Nrf2 null, Nrf2flox/flox, and Nrf2Δ/Δ; SPC-Cre) were administered a thoracic dose of 12Gy and allowed to recover for 250 days. Whole slide digital and confocal microscopy imaging of H&E, Masson's trichrome and immunostaining were used to assess tissue remodeling, collagen deposition and cell renewal/mobilization during the regenerative process. Histological assessment of irradiated, fibrotic wild type lung revealed significant loss of alveolar type 2 cells 250 days after irradiation. Type 2 cell loss and the corresponding development of fibrosis were enhanced in the Nrf2 null mouse. Yet, conditional deletion of Nrf2 in alveolar type 2 cells in irradiated lung did not impair type 2 cell survival nor yield an increased fibrotic phenotype. Instead, radiation-induced ΔNp63 stem/progenitor cell mobilization was inhibited in the Nrf2 null mouse while the propensity for radiation-induced myofibroblasts derived from alveolar type 2 cells was magnified. In summary, these results indicate that Nrf2 is an important regulator of irradiated lung's capacity to maintain alveolar type 2 cells, whose injury can initiate a fibrotic phenotype. Loss of Nrf2 inhibits ΔNp63 stem/progenitor mobilization, a key event for reconstitution of injured lung, while promoting a myofibroblast phenotype that is central for fibrosis.


Subject(s)
Epithelial Cells/radiation effects , NF-E2-Related Factor 2/genetics , Phosphoproteins/genetics , Pulmonary Fibrosis/genetics , Respiratory Mucosa/radiation effects , Trans-Activators/genetics , X-Rays/adverse effects , Animals , Cell Proliferation/radiation effects , Cell Survival/radiation effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Gene Expression Regulation , Hematopoietic Stem Cell Mobilization , Lung/metabolism , Lung/pathology , Lung/radiation effects , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Myofibroblasts/metabolism , Myofibroblasts/pathology , Myofibroblasts/radiation effects , NF-E2-Related Factor 2/deficiency , Phosphoproteins/metabolism , Pulmonary Fibrosis/etiology , Pulmonary Fibrosis/metabolism , Pulmonary Fibrosis/pathology , Respiratory Mucosa/metabolism , Respiratory Mucosa/pathology , Signal Transduction , Stem Cells/metabolism , Stem Cells/pathology , Stem Cells/radiation effects , Thorax , Trans-Activators/metabolism
12.
Oncotarget ; 7(47): 77926-77936, 2016 Nov 22.
Article in English | MEDLINE | ID: mdl-27788492

ABSTRACT

The goal of this investigation was to clarify the question of whether targeting Enox1 in tumor stroma would synergistically enhance the survival of tumor-bearing mice treated with fractionated radiotherapy. Enox1, a NADH oxidase, is expressed in tumor vasculature and stroma. However, it is not expressed in many tumor types, including HT-29 colorectal carcinoma cells. Pharmacological inhibition of Enox1 in endothelial cells inhibited repair of DNA double strand breaks, as measured by γH2AX and 53BP1 foci formation, as well as neutral comet assays. For 4 consecutive days athymic mice bearing HT-29 hindlimb xenografts were injected with a small molecule inhibitor of Enox1 or solvent control. Tumors were then administered 2 Gy of x-rays. On day 5 tumors were administered a single 'top-up' fraction of 30 Gy, the purpose of which was to amplify intrinsic differences in the radiation fractionation regimen produced by Enox1 targeting. Pharmacological targeting of Enox1 resulted in 80% of the tumor-bearing mice surviving at 90 days compared to only 40% of tumor-bearing mice treated with solvent control. The increase in survival was not a consequence of reoxygenation, as measured by pimonidazole immunostaining. These results are interpreted to indicate that targeting of Enox1 in tumor stroma significantly enhances the effectiveness of 2 Gy fractionated radiotherapy and identifies Enox1 as a potential therapeutic target.


Subject(s)
Colorectal Neoplasms/radiotherapy , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Animals , Cell Line, Tumor , Colorectal Neoplasms/enzymology , Colorectal Neoplasms/pathology , Dose Fractionation, Radiation , HT29 Cells , Humans , Mice , Mice, Nude , Molecular Targeted Therapy , NADH, NADPH Oxidoreductases/metabolism , Radiation-Sensitizing Agents/pharmacology , Xenograft Model Antitumor Assays
13.
Sci Rep ; 6: 24919, 2016 04 27.
Article in English | MEDLINE | ID: mdl-27118599

ABSTRACT

Protein lysine modification by γ-ketoaldehyde isomers derived from arachidonic acid, termed isolevuglandins (IsoLGs), is emerging as a mechanistic link between pathogenic reactive oxygen species and disease progression. However, the questions of whether covalent modification of proteins by IsoLGs are subject to genetic regulation and the identity of IsoLG-modified proteins remain unclear. Herein we show that Nrf2 and Nox2 are key regulators of IsoLG modification in pulmonary tissue and report on the identity of proteins analyzed by LC-MS following immunoaffinity purification of IsoLG-modified proteins. Gene ontology analysis revealed that proteins in numerous cellular pathways are susceptible to IsoLG modification. Although cells tolerate basal levels of modification, exceeding them induces apoptosis. We found prominent modification in a murine model of radiation-induced pulmonary fibrosis and in idiopathic pulmonary fibrosis, two diseases considered to be promoted by gene-regulated oxidant stress. Based on these results we hypothesize that IsoLG modification is a hitherto unrecognized sequelae that contributes to radiation-induced pulmonary injury and IPF.


Subject(s)
Lung/chemistry , Lung/metabolism , Protein Processing, Post-Translational , Proteome/analysis , Pulmonary Fibrosis/pathology , Animals , Chromatography, Liquid , Disease Models, Animal , Mass Spectrometry , Mice
14.
Bioorg Med Chem ; 23(22): 7226-33, 2015 Nov 15.
Article in English | MEDLINE | ID: mdl-26602084

ABSTRACT

In the present study, we have designed and synthesized a series of 1-benzyl-2-methyl-3-indolylmethylene barbituric acid analogs (7a-7h) and 1-benzyl-2-methyl-3-indolylmethylene thiobarbituric acid analogs (7 i-7 l) as nucleophosmin 1 (NPM1) inhibitors and have evaluated them for their anti-cancer activity against a panel of 60 different human cancer cell lines. Among these analogs 7 i, 7 j, and 7 k demonstrated potent growth inhibitory effects in various cancer cell types with GI50 values <2 µM. Compound 7 k exhibited growth inhibitory effects on a sub-panel of six leukemia cell lines with GI50 values in the range 0.22-0.35 µM. Analog 7 i also exhibited GI50 values <0.35 µM against three of the leukemia cell lines in the sub-panel. Analogs 7 i, 7 j, 7 k and 7 l were also evaluated against the mutant NPM1 expressing OCI-AML3 cell line and compounds 7 k and 7 l were found to cause dose-dependent apoptosis (AP50 = 1.75 µM and 3.3 µM, respectively). Compound 7k also exhibited potent growth inhibition against a wide variety of solid tumor cell lines: that is, A498 renal cancer (GI50 = 0.19 µM), HOP-92 and NCI-H522 lung cancer (GI50 = 0.25 µM), COLO 205 and HCT-116 colon cancer (GI50 = 0.20 and 0.26 µM, respectively), CNS cancer SF-539 (GI50 = 0.22 µM), melanoma MDA-MB-435 (GI50 = 0.22 µM), and breast cancer HS 578T (GI50 = 0.22 µM) cell lines. Molecular docking studies suggest that compounds 7 k and 7 l exert their anti-leukemic activity by binding to a pocket in the central channel of the NPM1 pentameric structure. These results indicate that the small molecule inhibitors 7 i, 7 j, 7 k, and 7 l could be potentially developed into anti-NPM1 drugs for the treatment of a variety of hematologic malignancies and solid tumors.


Subject(s)
Antineoplastic Agents/chemistry , Barbiturates/chemistry , Nuclear Proteins/antagonists & inhibitors , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Barbiturates/chemical synthesis , Barbiturates/pharmacology , Binding Sites , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Screening Assays, Antitumor , Humans , Molecular Docking Simulation , Nuclear Proteins/metabolism , Nucleophosmin , Protein Structure, Tertiary
15.
Free Radic Biol Med ; 88(Pt B): 268-274, 2015 Nov.
Article in English | MEDLINE | ID: mdl-25975985

ABSTRACT

Nrf2 is a transcription factor that promotes antioxidant and drug-metabolizing gene expression. It also regulates the transcription of genes involved in carbohydrate and lipid metabolism, NADPH regeneration, and heme and iron metabolism, as well as proteasome metabolism. Emerging research has identified Nrf2 as a critical factor for promoting survival of mammalian cells subjected to ionizing radiation. At a mechanistic level, Nrf2 promotes the repair of DNA damage and drives detoxification of superoxide that is generated hours to days after irradiation. This review summarizes research in these areas and discusses targeting of Nrf2 in radiation-resistant cancer and Nrf2׳s role in mitigating acute radiation syndrome.


Subject(s)
Cell Survival/physiology , DNA Repair/physiology , NF-E2-Related Factor 2/metabolism , Radiation Tolerance/physiology , Radiation, Ionizing , Animals , Cell Survival/radiation effects , Humans
16.
Bioorg Med Chem ; 23(13): 3681-6, 2015 Jul 01.
Article in English | MEDLINE | ID: mdl-25922180

ABSTRACT

A series of indole analogs that are synthesized using the scaffold of a potent radiosensitizer, YTR107, were tested for their ability to alter the solubility of phosphorylated nucleophosmin 1 (pNPM1). NPM1 is critical for DNA double strand break (DSB) repair. In response to formation of DNA DSBs, phosphorylated T199 NPM1 binds to ubiquitinated chromatin, in a RNF8/RNF168-dependent manner, forming irradiation-induced foci (IRIF) that promote repair of DNA DSBs. A Western blot assay was developed using lead molecule, YTR107, for the purpose of screening newly synthesized molecules that target pNPM1 in irradiated cells. A colony formation assay was used to demonstrate the radiosensitization properties of the compounds. Compounds that enhanced the extractability of pNPM1 upon radiation treatment possessed radiosensitization properties.


Subject(s)
Barbiturates/pharmacology , Blotting, Western/methods , DNA Breaks, Double-Stranded/radiation effects , Indoles/pharmacology , Nuclear Proteins/isolation & purification , Radiation-Sensitizing Agents/pharmacology , Cell Line , Cell Line, Tumor , Cell Nucleus/chemistry , Cell Nucleus/metabolism , Cell Nucleus/radiation effects , Centrosome/chemistry , Centrosome/metabolism , Centrosome/radiation effects , Chromatin/chemistry , Chromatin/metabolism , Chromatin/radiation effects , Fibroblasts/cytology , Fibroblasts/metabolism , Fibroblasts/radiation effects , Gamma Rays , Humans , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Nucleophosmin , Phosphorylation , Tumor Stem Cell Assay , Ubiquitin/genetics , Ubiquitin/metabolism
17.
Int J Radiat Oncol Biol Phys ; 89(5): 1106-1114, 2014 Aug 01.
Article in English | MEDLINE | ID: mdl-25035215

ABSTRACT

PURPOSE: To test the hypothesis that small molecule targeting of nucleophosmin 1 (NPM1) represents a rational approach for radiosensitization. METHODS AND MATERIALS: Wilde-type and NPM1-deficient mouse embryo fibroblasts (MEFs) were used to determine whether radiosensitization produced by the small molecule YTR107 was NPM1 dependent. The stress response to ionizing radiation was assessed by quantifying pNPM1, γH2AX, and Rad51 foci, neutral comet tail moment, and colony formation. NPM1 levels in a human-derived non-small-cell lung cancer (NSCLC) tissue microarray (TMA) were determined by immunohistochemistry. YTR107-mediated radiosensitization was assessed in NSCLC cell lines and xenografts. RESULTS: Use of NPM1-null MEFs demonstrated that NPM1 is critical for DNA double- strand break (DSB) repair, that loss of NPM1 increases radiation sensitivity, and that YTR107-mediated radiosensitization is NPM1 dependent. YTR107 was shown to inhibit NPM1 oligomerization and impair formation of pNPM1 irradiation-induced foci that colocalized with γH2AX foci. Analysis of the TMA demonstrated that NPM1 is overexpressed in subsets of NSCLC. YTR107 inhibited DNA DSB repair and radiosensitized NSCLC lines and xenografts. CONCLUSIONS: These data demonstrate that YTR107-mediated targeting of NPM1 impairs DNA DSB repair, an event that increases radiation sensitivity.


Subject(s)
Barbiturates/pharmacology , DNA Breaks, Double-Stranded , DNA Repair/drug effects , Indoles/pharmacology , Molecular Targeted Therapy/methods , Neoplasm Proteins/drug effects , Nuclear Proteins/drug effects , Radiation Tolerance , Radiation-Sensitizing Agents/pharmacology , Animals , Carcinoma, Non-Small-Cell Lung/chemistry , Carcinoma, Non-Small-Cell Lung/radiotherapy , Fibroblasts/chemistry , Fibroblasts/radiation effects , Histones/analysis , Humans , Lung Neoplasms/chemistry , Lung Neoplasms/radiotherapy , Mice , Mice, Inbred C57BL , Mice, Nude , Neoplasm Proteins/analysis , Neoplasm Proteins/deficiency , Neoplasm Proteins/physiology , Nuclear Proteins/analysis , Nuclear Proteins/deficiency , Nuclear Proteins/physiology , Nucleophosmin , Rad51 Recombinase/analysis , Tissue Array Analysis , Tumor Cells, Cultured
18.
Cancer Res ; 74(1): 38-43, 2014 Jan 01.
Article in English | MEDLINE | ID: mdl-24247717

ABSTRACT

ENOX1 is a highly conserved NADH oxidase that helps to regulate intracellular nicotinamide adenine dinucleotide levels in many cell types, including endothelial cells. Pharmacologic and RNA interference (RNAi)-mediated suppression of ENOX1 impairs surrogate markers of tumor angiogenesis/vasculogenesis, providing support for the concept that ENOX1 represents an antiangiogenic druggable target. However, direct genetic evidence that demonstrates a role for ENOX1 in vascular development is lacking. In this study, we exploited a zebrafish embryonic model of development to address this question. Whole-mount in situ hybridization coupled with immunofluorescence performed on zebrafish embryos demonstrate that enox1 message and translated protein are expressed in most tissues, and its expression is enriched in blood vessels and heart. Morpholino-mediated suppression of Enox1 in Tg(fli1-eGFP) and Tg(flk1-eGFP) zebrafish embryos significantly impairs the development of vasculature and blood circulation. Using in vivo multiphoton microscopy, we show that morpholino-mediated knockdown of enox1 increases NADH levels, consistent with loss of enzyme. VJ115 is a small-molecule inhibitor of Enox1's oxidase activity shown to increase intracellular NADH in endothelial cells; we used VJ115 to determine if the oxidase activity was crucial for vascular development. We found that VJ115 suppressed vasculogenesis in Tg(fli1-eGFP) embryos and impaired circulation. Previously, it was shown that suppression of ENOX1 radiosensitizes proliferating tumor vasculature, a consequence of enhanced endothelial cell apoptosis. Thus, our current findings, coupled with previous research, support the hypothesis that ENOX1 represents a potential cancer therapy target, one that combines molecular targeting with cytotoxic sensitization.


Subject(s)
Endothelium, Vascular/embryology , Endothelium, Vascular/growth & development , Multienzyme Complexes/physiology , NADH, NADPH Oxidoreductases/physiology , Animals , Animals, Genetically Modified , Endothelium, Vascular/enzymology , Multienzyme Complexes/genetics , Multienzyme Complexes/metabolism , NADH, NADPH Oxidoreductases/genetics , NADH, NADPH Oxidoreductases/metabolism , Neovascularization, Physiologic/physiology , Zebrafish
19.
Ann Hepatol ; 12(6): 952-8, 2013.
Article in English | MEDLINE | ID: mdl-24114826

ABSTRACT

BACKGROUND AND AIMS: Most portal vein thromboses (PVT) in cirrhotics are discovered incidentally. While case series demonstrate improved portal vein patency with anti-coagulation, there is little information on impact of PVT on morbidity and mortality. This study aimed to compare morbidity and mortality in cirrhotics with untreated PVT with those without PVT. MATERIAL AND METHODS: Cirrhotics evaluated for orthotopic liver transplant in a single large transplant center were prospectively followed. Subjects had contrast CT or MRI at initial evaluation and serial imaging every 6 months until transplantation, removal from the list or death. Univariate and multivariate Cox regression analysis were used to assess associations between new PVT and factors of interest. RESULTS: Of the 290 prospectively followed cirrhotics who met inclusion criteria, PVT was detected in 70 (24.1%)-47 had PVT at the time of initial evaluation and 23 developed one during the pre-transplant study period. A third of the patients with PVT had re-canalization or spontaneous resolution of thrombus while awaiting transplantation. There was no difference in the pre or posttransplant mortality between cirrhotics with and without PVT. CONCLUSION: Cirrhotics with untreated PVT fared equally well as those without PVT before and after transplantation. Further studies with larger numbers of patients are needed to determine if anticoagulation therapy truly improves outcomes in cirrhotics with portal vein thrombosis.


Subject(s)
Liver Cirrhosis/surgery , Liver Transplantation , Portal Vein , Venous Thrombosis/epidemiology , Aged , Chi-Square Distribution , Female , Humans , Incidence , Kaplan-Meier Estimate , Liver Cirrhosis/diagnosis , Liver Cirrhosis/mortality , Liver Transplantation/adverse effects , Liver Transplantation/mortality , Magnetic Resonance Angiography , Male , Middle Aged , Multivariate Analysis , Ohio/epidemiology , Phlebography/methods , Portal Vein/diagnostic imaging , Portal Vein/pathology , Proportional Hazards Models , Prospective Studies , Remission, Spontaneous , Risk Factors , Time Factors , Tomography, X-Ray Computed , Treatment Outcome , Venous Thrombosis/diagnosis , Venous Thrombosis/mortality , Waiting Lists
20.
Invest New Drugs ; 31(3): 535-44, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23054211

ABSTRACT

Targeting tumor vasculature represents a rational strategy for controlling cancer. (Z)-(+/-)-2-(1-benzylindol-3-ylmethylene)-1-azabicyclo[2.2.2]octan-3-ol (denoted VJ115) is a novel chemical entity that inhibits the enzyme ENOX1, a NADH oxidase. Genetic and small molecule inhibition of ENOX1 inhibits endothelial cell tubule formation and tumor-mediated neo-angiogenesis. Inhibition of ENOX1 radiosensitizes tumor vasculature, a consequence of enhanced apoptosis. However, the molecular mechanisms underlying these observations are not well understood. Herein, we mechanistically link ENOX1-mediated regulation of cellular NADH concentrations with proteomics profiling of endothelial cell protein expression following exposure to VJ115. Pathway Studios network analysis of potential effector molecules identified by the proteomics profiling indicated that a VJ115 exposure capable of altering intracellular NADH concentrations impacted proteins involved in cytoskeletal reorganization. The analysis was validated using RT-PCR and immunoblotting of selected proteins. RNAi knockdown of ENOX1 was shown to suppress expression of stathmin and lamin A/C, proteins identified by the proteomics analysis to be suppressed upon VJ115 exposure. These data support the hypothesis that VJ115 inhibition of ENOX1 can impact expression of proteins involved in cytoskeletal reorganization and support a hypothesis in which ENOX1 activity links elevated cellular NADH concentrations with cytoskeletal reorganization and angiogenesis.


Subject(s)
Angiogenesis Inhibitors/pharmacology , Cytoskeletal Proteins/metabolism , Indoles/pharmacology , NADH, NADPH Oxidoreductases/antagonists & inhibitors , Quinuclidines/pharmacology , Cells, Cultured , Human Umbilical Vein Endothelial Cells/drug effects , Human Umbilical Vein Endothelial Cells/metabolism , Humans , NAD/metabolism , Proteomics
SELECTION OF CITATIONS
SEARCH DETAIL
...