Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 16 de 16
Filter
Add more filters










Publication year range
1.
Photochem Photobiol ; 96(5): 1014-1031, 2020 09.
Article in English | MEDLINE | ID: mdl-32221980

ABSTRACT

Psoralen is a furocoumarin natural product that intercalates within DNA and forms covalent adducts when activated by ultraviolet radiation. It is well known that this property contributes to psoralen's clinical efficacy in several disease contexts, which include vitiligo, psoriasis, graft-versus-host disease and cutaneous T-cell lymphoma. Given the therapeutic relevance of psoralen and its derivatives, we attempted to synthesize psoralens with even greater potency. In this study, we report a library of 73 novel psoralens, the largest collection of its kind. When screened for the ability to reduce cell proliferation, we identified two derivatives even more cytotoxic than 4'-aminomethyl-4,5',8-trimethylpsoralen (AMT), one of the most potent psoralens identified to date. Using MALDI-TOF MS, we studied the DNA adduct formation for a subset of novel psoralens and found that in most cases enhanced DNA binding correlated well with cytotoxicity. Generally, our most potent derivatives contain positively charged substituents, which we believe increase DNA affinity and enhance psoralen intercalation. Thus, we provide a rational approach to guide efforts toward further optimizing psoralens to fully capitalize on this drug class' therapeutic potential. Finally, the structure-activity insights we have gained shed light on several opportunities to study currently underappreciated aspects of psoralen's mechanism.


Subject(s)
DNA/drug effects , Furocoumarins/pharmacology , Animals , Cell Line, Tumor , DNA/chemistry , DNA Adducts , Furocoumarins/chemistry , Mice , Spectrometry, Mass, Matrix-Assisted Laser Desorption-Ionization , Structure-Activity Relationship , Ultraviolet Rays
2.
Radiat Res ; 184(5): 456-69, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26488758

ABSTRACT

Although a standardized approach to radiotherapy has been used to treat breast cancer, regardless of subtype (e.g., luminal, basal), recent clinical data suggest that radiation response may vary significantly among subtypes. We hypothesized that this clinical variability may be due, in part, to differences in cellular radiation response. In this study, we utilized RNA samples for microarray analysis from two sources: 1. Paired pre- and postirradiation breast tumor tissue from 32 early-stage breast cancer patients treated in our unique preoperative radiation Phase I trial; and 2. Sixteen biologically diverse breast tumor cell lines exposed to 0 and 5 Gy irradiation. The transcriptome response to radiation exposure was derived by comparing gene expression in samples before and after irradiation. Genes with the highest coefficient of variation were selected for further evaluation and validated at the RNA and protein level. Gene editing and agonistic antibody treatment were performed to assess the impact of gene modulation on radiation response. Gene expression in our cohort of luminal breast cancer patients was distinctly different before and after irradiation. Further, two distinct patterns of gene expression were observed in our biologically diverse group of breast cancer cell lines pre- versus postirradiation. Cell lines that showed significant change after irradiation were largely luminal subtype, while gene expression in the basal and HER2+ cell lines was minimally impacted. The 100 genes with the most significant response to radiation in patients were identified and analyzed for differential patterns of expression in the radiation-responsive versus nonresponsive cell lines. Fourteen genes were identified as significant, including FAS, a member of the tumor necrosis factor receptor family known to play a critical role in programed cell death. Modulation of FAS in breast cancer cell lines altered radiation response phenotype and enhanced radiation sensitivity in radioresistant basal cell lines. Our findings suggest that cell-type-specific, radiation-induced FAS contributes to subtype-specific breast cancer radiation response and that activation of FAS pathways may be exploited for biologically tailored radiotherapy.


Subject(s)
Biomarkers, Tumor/genetics , Breast Neoplasms/genetics , Breast Neoplasms/radiotherapy , Molecular Targeted Therapy , Radiation Tolerance/genetics , fas Receptor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/pathology , Cell Line, Tumor , Female , Genes, p53/genetics , Humans , Middle Aged , Oligonucleotide Array Sequence Analysis , Phenotype , Protein Transport/radiation effects , Transcriptional Activation/radiation effects , Transcriptome/radiation effects , fas Receptor/metabolism
3.
AJR Am J Roentgenol ; 201(5): W698-706, 2013 Nov.
Article in English | MEDLINE | ID: mdl-24147499

ABSTRACT

OBJECTIVE: Hypoxia is a characteristic of many tumors and portends a worse prognosis in lung, cervical, prostate, and rectal cancers. Unlike the others, lung cancers present a unique challenge in measuring hypoxia, with invasive biopsies and higher rates of complications. Noninvasive imaging studies detecting hypoxia using isotopes of copper-diacetyl-bis(N4-methylthiosemicarbazone) ((62)Cu-ATSM) have predicted prognosis and treatment outcomes in some small feasibility trials. These images, however, may not identify all areas of hypoxia. Hence, we hypothesize that the addition of another PET imaging agent, copper-pyruvaldehyde-bis(N4-methylthiosemicarbazone) ((62)Cu-PTSM), which can detect areas of perfusion, can augment the information obtained in (62)Cu-ATSM PET scans. SUBJECTS AND METHODS: To characterize tumors on the basis of both perfusion and hypoxia, 10 patients were studied using both (62)Cu-ATSM and (62)Cu-PTSM PET scans. In addition, proteomic arrays looking at specific proangiogenic, survival, and proinflammatory targets were assessed. RESULTS: Six of 10 patients had evaluable PET scans. Our initial experience of characterizing lung tumor hypoxia using (62)Cu-ATSM and (62)Cu-PTSM PET scans showed that visualization of areas with hypoxia normalized for perfusion is feasible. All studied tumors exhibited some hypoxia. Despite the small sample size, a positive relationship was noted between epidermal growth factor levels and (62)Cu-ATSM-detected hypoxia. CONCLUSION: This initial series of (62)Cu-ATSM and (62)Cu-PTSM PET scans shows that evaluating lung masses by visualizing hypoxia and perfusion is a feasible and novel technique to provide more information. Further investigation is warranted to assess the potential role of (62)Cu-ATSM and (62)Cu-PTSM PET techniques combined with proteomics as alternatives to invasive biopsy techniques in clinical care.


Subject(s)
Lung Neoplasms/diagnostic imaging , Organometallic Compounds , Thiosemicarbazones , Aged , Coordination Complexes , Cytokines/blood , Female , Fluorodeoxyglucose F18 , Humans , Hypoxia , Lung Neoplasms/pathology , Male , Middle Aged , Neoplasm Staging , Positron-Emission Tomography/methods , Radiopharmaceuticals , Survival Rate
4.
Cancer Prev Res (Phila) ; 6(9): 925-37, 2013 Sep.
Article in English | MEDLINE | ID: mdl-23842792

ABSTRACT

Aerobic exercise training (AET) is an effective adjunct therapy to attenuate the adverse side-effects of adjuvant chemotherapy in women with early breast cancer. Whether AET interacts with the antitumor efficacy of chemotherapy has received scant attention. We carried out a pilot study to explore the effects of AET in combination with neoadjuvant doxorubicin-cyclophosphamide (AC+AET), relative to AC alone, on: (i) host physiology [exercise capacity (VO2 peak), brachial artery flow-mediated dilation (BA-FMD)], (ii) host-related circulating factors [circulating endothelial progenitor cells (CEP) cytokines and angiogenic factors (CAF)], and (iii) tumor phenotype [tumor blood flow ((15)O-water PET), tissue markers (hypoxia and proliferation), and gene expression] in 20 women with operable breast cancer. AET consisted of three supervised cycle ergometry sessions/week at 60% to 100% of VO2 peak, 30 to 45 min/session, for 12 weeks. There was significant time × group interactions for VO2 peak and BA-FMD, favoring the AC+AET group (P < 0.001 and P = 0.07, respectively). These changes were accompanied by significant time × group interactions in CEPs and select CAFs [placenta growth factor, interleukin (IL)-1ß, and IL-2], also favoring the AC+AET group (P < 0.05). (15)O-water positron emission tomography (PET) imaging revealed a 38% decrease in tumor blood flow in the AC+AET group. There were no differences in any tumor tissue markers (P > 0.05). Whole-genome microarray tumor analysis revealed significant differential modulation of 57 pathways (P < 0.01), including many that converge on NF-κB. Data from this exploratory study provide initial evidence that AET can modulate several host- and tumor-related pathways during standard chemotherapy. The biologic and clinical implications remain to be determined.


Subject(s)
Adenocarcinoma/therapy , Angiogenesis Inducing Agents/blood , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/therapy , Carcinoma, Ductal, Breast/therapy , Exercise Therapy , Neoadjuvant Therapy , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Biomarkers, Tumor/genetics , Biomarkers, Tumor/metabolism , Breast Neoplasms/drug therapy , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/drug therapy , Carcinoma, Ductal, Breast/pathology , Chemotherapy, Adjuvant , Combined Modality Therapy , Cyclophosphamide/administration & dosage , Doxorubicin/administration & dosage , Endothelium, Vascular/metabolism , Endothelium, Vascular/pathology , Female , Gene Expression Profiling , Humans , Immunoenzyme Techniques , Middle Aged , Neoplasm Staging , Neovascularization, Pathologic , Oligonucleotide Array Sequence Analysis , Prognosis , RNA, Messenger/genetics , Real-Time Polymerase Chain Reaction , Reverse Transcriptase Polymerase Chain Reaction , Stem Cells/metabolism , Stem Cells/pathology
5.
PLoS One ; 8(1): e52543, 2013.
Article in English | MEDLINE | ID: mdl-23326340

ABSTRACT

Resistance of hypoxic solid tumor niches to chemotherapy and radiotherapy remains a major scientific challenge that calls for conceptually new approaches. Here we exploit a hitherto unrecognized ability of sickled erythrocytes (SSRBCs) but not normal RBCs (NLRBCs) to selectively target hypoxic tumor vascular microenviroment and induce diffuse vaso-occlusion. Within minutes after injection SSRBCs, but not NLRBCs, home and adhere to hypoxic 4T1 tumor vasculature with hemoglobin saturation levels at or below 10% that are distributed over 70% of the tumor space. The bound SSRBCs thereupon form microaggregates that obstruct/occlude up to 88% of tumor microvessels. Importantly, SSRBCs, but not normal RBCs, combined with exogenous prooxidant zinc protoporphyrin (ZnPP) induce a potent tumoricidal response via a mutual potentiating mechanism. In a clonogenic tumor cell survival assay, SSRBC surrogate hemin, along with H(2)O(2) and ZnPP demonstrate a similar mutual potentiation and tumoricidal effect. In contrast to existing treatments directed only to the hypoxic tumor cell, the present approach targets the hypoxic tumor vascular environment and induces injury to both tumor microvessels and tumor cells using intrinsic SSRBC-derived oxidants and locally generated ROS. Thus, the SSRBC appears to be a potent new tool for treatment of hypoxic solid tumors, which are notable for their resistance to existing cancer treatments.


Subject(s)
Cytotoxicity, Immunologic/immunology , Erythrocytes, Abnormal/immunology , Neoplasms, Experimental/immunology , Neovascularization, Pathologic/immunology , Anemia, Sickle Cell/blood , Anemia, Sickle Cell/immunology , Animals , Blotting, Western , Cell Line, Tumor , Combined Modality Therapy , Erythrocytes, Abnormal/metabolism , Erythrocytes, Abnormal/transplantation , Female , Heme Oxygenase-1/metabolism , Hemin/metabolism , Humans , Hydrogen Peroxide/metabolism , Hypoxia , Immunotherapy, Adoptive , Membrane Proteins/metabolism , Mice , Mice, Nude , Microscopy, Fluorescence , Neoplasms, Experimental/blood supply , Neoplasms, Experimental/pathology , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/therapy , Protoporphyrins/pharmacology , Reactive Oxygen Species/immunology , Reactive Oxygen Species/metabolism , Tumor Microenvironment/drug effects , Tumor Microenvironment/immunology
6.
J Appl Physiol (1985) ; 113(2): 263-72, 2012 Jul.
Article in English | MEDLINE | ID: mdl-22604887

ABSTRACT

The purpose of this study is to investigate the effects of exercise on cancer progression, metastasis, and underlying mechanisms in an orthotopic model of murine prostate cancer. C57BL/6 male mice (6-8 wk of age) were orthotopically injected with transgenic adenocarcinoma of mouse prostate C-1 cells (5 × 10(5)) and randomly assigned to exercise (n = 28) or a non-intervention control (n = 31) groups. The exercise group was given voluntary access to a wheel 24 h/day for the duration of the study. Four mice per group were serially killed on days 14, 31, and 36; the remaining 38 mice (exercise, n = 18; control, n = 20) were killed on day 53. Before death, MRI was performed to assess tumor blood perfusion. Primary tumor growth rate was comparable between groups, but expression of prometastatic genes was significantly modulated in exercising animals with a shift toward reduced metastasis. Exercise was associated with increased activity of protein kinases within the MEK/MAPK and PI3K/mTOR signaling cascades with subsequent increased intratumoral protein levels of HIF-1α and VEGF. This was associated with improved tumor vascularization. Multiplex ELISAs revealed distinct reductions in plasma concentrations of several angiogenic cytokines in the exercise group, which was associated with increased expression of angiogenic and metabolic genes in the skeletal muscle. Exercise-induced stabilization of HIF-1α and subsequent upregulation of VEGF was associated with "productive" tumor vascularization with a shift toward suppressed metastasis in an orthotopic model of prostate cancer.


Subject(s)
Adenocarcinoma/physiopathology , Adenocarcinoma/secondary , Cytokines/blood , Exercise Therapy/methods , Neovascularization, Pathologic/physiopathology , Physical Conditioning, Animal/methods , Prostatic Neoplasms/physiopathology , Adenocarcinoma/prevention & control , Animals , Male , Mice , Mice, Inbred C57BL , Neoplasms, Experimental , Neovascularization, Pathologic/prevention & control , Prostatic Neoplasms/prevention & control , Prostatic Neoplasms/secondary , Treatment Outcome
7.
Free Radic Res ; 45(11-12): 1289-306, 2011 Nov.
Article in English | MEDLINE | ID: mdl-21859376

ABSTRACT

Due to the ability to easily accept and donate electrons Mn(III)N-alkylpyridylporphyrins (MnPs) can dismute O(2)(·-), reduce peroxynitrite, but also generate reactive species and behave as pro-oxidants if conditions favour such action. Herein two ortho isomers, MnTE-2-PyP(5+), MnTnHex-2-PyP(5+), and a meta isomer MnTnHex-3-PyP(5+), which differ greatly with regard to their metal-centered reduction potential, E(1/2) (Mn(III)P/Mn(II)P) and lipophilicity, were explored. Employing Mn(III)P/Mn(II)P redox system for coupling with ascorbate, these MnPs catalyze ascorbate oxidation and thus peroxide production. Consequently, cancer oxidative burden may be enhanced, which in turn would suppress its growth. Cytotoxic effects on Caco-2, Hela, 4T1, HCT116 and SUM149 were studied. When combined with ascorbate, MnPs killed cancer cells via peroxide produced outside of the cell. MnTE-2-PyP(5+) was the most efficacious catalyst for peroxide production, while MnTnHex-3-PyP(5+) is most prone to oxidative degradation with H(2) , and thus the least efficacious. A 4T1 breast cancer mouse study of limited scope and success was conducted. The tumour oxidative stress was enhanced and its microvessel density reduced when mice were treated either with ascorbate or MnP/ascorbate; the trend towards tumour growth suppression was detected.


Subject(s)
Antineoplastic Agents/pharmacology , Ascorbic Acid/pharmacology , Metalloporphyrins/pharmacology , Reactive Oxygen Species/pharmacology , Reducing Agents/pharmacology , Animals , Antineoplastic Agents/chemistry , Ascorbic Acid/chemistry , Caco-2 Cells , Catalysis , Female , HCT116 Cells , HeLa Cells , Humans , Isomerism , Metalloporphyrins/chemistry , Mice , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Peroxides/metabolism , Reactive Oxygen Species/metabolism , Reducing Agents/chemistry
8.
J Clin Oncol ; 29(21): 2918-23, 2011 Jul 20.
Article in English | MEDLINE | ID: mdl-21690470

ABSTRACT

PURPOSE: Identifying strong markers of prognosis are critical to optimize treatment and survival outcomes in patients with malignant recurrent glioma. We investigated the prognostic significance of exercise behavior and functional capacity in this population. PATIENTS AND METHODS: Using a prospective design, 243 patients with WHO grades 3 to 4 recurrent malignant glioma and Karnofsky performance status (KPS) ≥ 70 completed a self-administered questionnaire that assessed exercise behavior and performed a 6-minute walk test (6MWT) to assess functional capacity. Cox proportional models were used to estimate the risk of all-cause mortality according to 6MWT distance (6MWD; < 390 meters, 390-489 meters, > 489 meters) and exercise behavior (metabolic equivalent [MET] -h/wk) adjusted for KPS and other important clinical factors. RESULTS: Median follow-up was 27.43 months. During this period, 149 deaths were recorded (61% of the total sample). Exercise behavior was an independent predictor of survival (P = .0081). Median survival was 13.03 months for patients reporting < 9 MET-h/wk relative to 21.84 months for those reporting ≥ 9 MET-h/wk. Exercise behavior added incremental prognostic value beyond that provided by KPS, age, sex, grade, and number of prior progressions (P < .001). Compared with patients reporting < 9 MET-h/wk, the adjusted hazard ratio for mortality was 0.64 (95% CI, 0.46 to 0.91) for patients reporting ≥ 9 MET-h/wk. Functional capacity was not an independent predictor of prognosis. CONCLUSION: Exercise behavior is a strong independent predictor of survival that provides incremental prognostic value to KPS as well as traditional markers of prognosis in malignant recurrent glioma.


Subject(s)
Brain Neoplasms/mortality , Exercise , Glioma/mortality , Health Behavior , Neoplasm Recurrence, Local , Walking , Adult , Aged , Brain Neoplasms/pathology , Brain Neoplasms/physiopathology , Brain Neoplasms/psychology , Brain Neoplasms/therapy , Exercise Test , Female , Glioma/pathology , Glioma/physiopathology , Glioma/psychology , Glioma/therapy , Humans , Karnofsky Performance Status , Male , Middle Aged , North Carolina , Predictive Value of Tests , Proportional Hazards Models , Prospective Studies , Risk Assessment , Risk Factors , Surveys and Questionnaires , Survival Analysis , Survival Rate , Time Factors , Treatment Outcome
9.
Clin Cancer Res ; 17(8): 2549-60, 2011 Apr 15.
Article in English | MEDLINE | ID: mdl-21292819

ABSTRACT

PURPOSE: While hyperthermia is an effective adjuvant treatment to radiotherapy, we do not completely understand the nature of the response heterogeneity. EXPERIMENTAL DESIGN: We performed gene expression analysis of 22 spontaneous canine sarcomas before and after the first hyperthermia treatment administered as an adjuvant to radiotherapy. In parallel, diffusion-weighted MRI (DWI) was done prior to the treatment course and at the end of therapy. RESULTS: From the integrative analysis of gene expression and DWI, we identified significant correlation between tumor responses with genes involved in VEGF signaling, telomerase, DNA repair, and inflammation. The treatment-induced changes in gene expression identified 2 distinct tumor subtypes with significant differences in their gene expression and treatment response, as defined by changes in DWI. The 2 tumor subtypes could also be readily identified by pretreatment gene expression. The tumor subtypes, with stronger expression response and DWI increase, had higher levels of HSP70, POT1, and centrosomal proteins, and lower levels of CD31, vWF, and transferrin. Such differential gene expression between the 2 subtypes was used to interrogate connectivity map and identify linkages to an HSP90 inhibitor, geldanamycin. We further validated the ability of geldanamycin to enhance cell killing of human tumor cells with hyperthermia and radiotherapy in clonogenic assays. CONCLUSIONS: To our knowledge, this is one of the first successful attempts to link changes in gene expression and functional imaging to understand the response heterogeneity and identify compounds enhancing thermoradiotherapy. This study also demonstrates the value of canine tumors to provide information generalizable to human tumors.


Subject(s)
Genomics/methods , Magnetic Resonance Imaging/methods , Sarcoma/genetics , Sarcoma/therapy , Animals , Cluster Analysis , Combined Modality Therapy , Dogs , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Humans , Hyperthermia, Induced , Oligonucleotide Array Sequence Analysis , Radiotherapy/methods , Reverse Transcriptase Polymerase Chain Reaction
10.
Free Radic Res ; 45(2): 188-200, 2011 Feb.
Article in English | MEDLINE | ID: mdl-20942564

ABSTRACT

In the cell Mn porphyrins (MnPs) likely couple with cellular reductants which results in a drop of total charge from 5+ to 4+ and dramatically increases their lipophilicity by up to three orders of magnitude depending upon the length of alkylpyridyl chains and type of isomer. The effects result from the interplay of solvation, lipophilicit and stericity. Impact of ascorbate on accumulation of MnPs was measured in E. coli and in Balb/C mouse tumours and muscle; for the latter measurements, the LC/ESI-MS/MS method was developed. Accumulation was significantly enhanced when MnPs were co-administered with ascorbate in both prokaryotic and eukaryotic systems. Further, MnTnHex-2-PyP(5+) accumulates 5-fold more in the tumour than in a muscle. Such data increase our understanding of MnPs cellular and sub-cellular accumulation and remarkable in vivo effects. The work is in progress to understand how coupling of MnPs with ascorbate affects their mechanism of action, in particular with respect to cancer therapy.


Subject(s)
Manganese/chemistry , Metalloporphyrins/pharmacokinetics , Molecular Mimicry , Superoxide Dismutase/pharmacokinetics , Animals , Ascorbic Acid/administration & dosage , Ascorbic Acid/therapeutic use , Biological Availability , Chromatography, Liquid , Escherichia coli/drug effects , Escherichia coli/metabolism , Female , Hydrophobic and Hydrophilic Interactions/drug effects , Ion Transport , Isomerism , Manganese/metabolism , Mass Spectrometry , Metalloporphyrins/metabolism , Metalloporphyrins/therapeutic use , Mice , Muscles/drug effects , Muscles/metabolism , Neoplasms/drug therapy , Neoplasms/metabolism , Oxidation-Reduction/drug effects , Solubility/drug effects , Static Electricity , Superoxide Dismutase/metabolism , Superoxide Dismutase/therapeutic use
11.
Cancer Res ; 68(22): 9323-30, 2008 Nov 15.
Article in English | MEDLINE | ID: mdl-19010906

ABSTRACT

Hypoxia is a dynamic feature of the tumor microenvironment that contributes to drug resistance and cancer progression. We previously showed that components of the unfolded protein response (UPR), elicited by endoplasmic reticulum (ER) stress, are also activated by hypoxia in vitro and in vivo animal and human patient tumors. Here, we report that ER stressors, such as thapsigargin or the clinically used proteasome inhibitor bortezomib, exhibit significantly higher cytotoxicity toward hypoxic compared with normoxic tumor cells, which is accompanied by enhanced activation of UPR effectors in vitro and UPR reporter activity in vivo. Treatment of cells with the translation inhibitor cycloheximide, which relieves ER load, ameliorated this enhanced cytotoxicity, indicating that the increased cytotoxicity is ER stress-dependent. The mode of cell death was cell type-dependent, because DLD1 colorectal carcinoma cells exhibited enhanced apoptosis, whereas HeLa cervical carcinoma cells activated autophagy, blocked apoptosis, and eventually led to necrosis. Pharmacologic or genetic ablation of autophagy increased the levels of apoptosis. These results show that hypoxic tumor cells, which are generally more resistant to genotoxic agents, are hypersensitive to proteasome inhibitors and suggest that combining bortezomib with therapies that target the normoxic fraction of human tumors can lead to more effective tumor control.


Subject(s)
Antineoplastic Agents/pharmacology , Boronic Acids/pharmacology , Endoplasmic Reticulum/drug effects , Protease Inhibitors/pharmacology , Pyrazines/pharmacology , Stress, Physiological/drug effects , Activating Transcription Factor 4/genetics , Apoptosis , Autophagy , Bortezomib , Cell Hypoxia , Cell Line, Tumor , DNA-Binding Proteins/genetics , Endoplasmic Reticulum/metabolism , Humans , Microtubule-Associated Proteins/metabolism , Necrosis , Protein Folding , Regulatory Factor X Transcription Factors , Signal Transduction , Thapsigargin/pharmacology , Transcription Factors/genetics
12.
Cancer Res ; 67(3): 1262-9, 2007 Feb 01.
Article in English | MEDLINE | ID: mdl-17283163

ABSTRACT

Fatty acid synthase (FAS), the cellular enzyme that synthesizes palmitate, is expressed at high levels in tumor cells and is vital for their survival. Through the synthesis of palmitate, FAS primarily drives the synthesis of phospholipids in tumor cells. In this study, we tested the hypothesis that the FAS inhibitors induce endoplasmic reticulum (ER) stress in tumor cells. Treatment of tumor cells with FAS inhibitors induces robust PERK-dependent phosphorylation of the translation initiation factor eIF2alpha and concomitant inhibition of protein synthesis. PERK-deficient transformed mouse embryonic fibroblasts and HT-29 colon carcinoma cells that express a dominant negative PERK (DeltaC-PERK) are hypersensitive to FAS inhibitor-induced cell death. Pharmacologic inhibition of FAS also induces the processing of X-box binding protein-1, indicating that the IRE1 arm of the ER stress response is activated when FAS is inhibited. Induction of ER stress is further confirmed by the increased expression of the ER stress-regulated genes CHOP, ATF4, and GRP78. FAS inhibitor-induced ER stress is activated prior to the detection of caspase 3 and PARP cleavage, primary indicators of cell death, whereas orlistat-induced cell death is rescued by coincubation with the global translation inhibitor cycloheximide. Lastly, FAS inhibitors cooperate with the ER stress inducer thapsigargin to enhance tumor cell killing. These results provide the first evidence that FAS inhibitors induce ER stress and establish an important mechanistic link between FAS activity and ER function.


Subject(s)
Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum/enzymology , Enzyme Inhibitors/pharmacology , Fatty Acid Synthases/antagonists & inhibitors , Prostatic Neoplasms/enzymology , Activating Transcription Factor 4/biosynthesis , Activating Transcription Factor 4/genetics , Animals , Cell Line, Tumor , DNA-Binding Proteins/biosynthesis , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Drug Interactions , Endoplasmic Reticulum/genetics , Endoplasmic Reticulum Chaperone BiP , Eukaryotic Initiation Factor-2B/metabolism , Fatty Acid Synthases/biosynthesis , HT29 Cells , HeLa Cells , Heat-Shock Proteins/biosynthesis , Heat-Shock Proteins/genetics , Humans , Lactones/pharmacology , Male , Mice , Molecular Chaperones/biosynthesis , Molecular Chaperones/genetics , Nuclear Proteins/biosynthesis , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Orlistat , Phosphorylation , Prostatic Neoplasms/drug therapy , Regulatory Factor X Transcription Factors , Thapsigargin/pharmacology , Transcription Factor CHOP/biosynthesis , Transcription Factor CHOP/genetics , Transcription Factors , eIF-2 Kinase/metabolism
13.
Cancer Biol Ther ; 5(7): 723-8, 2006 Jul.
Article in English | MEDLINE | ID: mdl-16861899

ABSTRACT

Hypoxia is a dynamic feature of the tumor microenvironment that contributes to cancer progression. In order to adapt and overcome hypoxic stress, tumor cells activate survival pathways that attempt to couple metabolic processes to reduced energy availability due to oxygen deprivation. While the hypoxia-inducible factors HIF-1 and HIF-2 are critical to the cellular response to hypoxia, HIF-independent processes are known to contribute to this adaptation. Recent evidence demonstrates that hypoxia activates components of the Unfolded Protein Response (UPR), a coordinated program that regulates cellular adaptation to increased levels of unfolded proteins in the endoplasmic reticulum (ER). Here we review the evidence implicating the ER kinase PERK, its downstream target translation initiation factor eIF2alpha, and the subsequent translational upregulation of the transcription factor ATF4 in this response. Not only are cells with compromised PERK-eIF2alpha-ATF4 signaling more sensitive to hypoxic stress in vitro but they also form tumors that grow slower in vivo with smaller hypoxic areas, indicating that the PERK-eIF2alpha-ATF4 pathway confers a survival advantage for tumor cells under hypoxia. These results, together with evidence for an involvement of other UPR pathways and ER stress proteins in hypoxia tolerance and tumor maintenance, point to a central role for UPR activation in tumor progression and suggest that this response may offer an attractive target for new anti-tumor modalities.


Subject(s)
Activating Transcription Factor 4/metabolism , Eukaryotic Initiation Factor-2/metabolism , Neoplasms/metabolism , Neoplasms/pathology , eIF-2 Kinase/metabolism , Animals , Cell Hypoxia , Endoplasmic Reticulum , Humans , Protein Folding , Signal Transduction
14.
EMBO J ; 24(19): 3470-81, 2005 Oct 05.
Article in English | MEDLINE | ID: mdl-16148948

ABSTRACT

Tumor cell adaptation to hypoxic stress is an important determinant of malignant progression. While much emphasis has been placed on the role of HIF-1 in this context, the role of additional mechanisms has not been adequately explored. Here we demonstrate that cells cultured under hypoxic/anoxic conditions and transformed cells in hypoxic areas of tumors activate a translational control program known as the integrated stress response (ISR), which adapts cells to endoplasmic reticulum (ER) stress. Inactivation of ISR signaling by mutations in the ER kinase PERK and the translation initiation factor eIF2alpha or by a dominant-negative PERK impairs cell survival under extreme hypoxia. Tumors derived from these mutant cell lines are smaller and exhibit higher levels of apoptosis in hypoxic areas compared to tumors with an intact ISR. Moreover, expression of the ISR targets ATF4 and CHOP was noted in hypoxic areas of human tumor biopsy samples. Collectively, these findings demonstrate that activation of the ISR is required for tumor cell adaptation to hypoxia, and suggest that this pathway is an attractive target for antitumor modalities.


Subject(s)
Endoplasmic Reticulum/physiology , Eukaryotic Initiation Factor-2/metabolism , Gene Expression Regulation, Neoplastic , Hypoxia/physiopathology , Neoplasms/physiopathology , Signal Transduction/physiology , Stress, Physiological/physiopathology , eIF-2 Kinase/metabolism , Activating Transcription Factor 4/metabolism , Animals , Apoptosis/genetics , Apoptosis/physiology , Cell Line, Tumor , Cell Survival/genetics , Cell Survival/physiology , Humans , Immunohistochemistry , Mice , Mice, Nude , Mutation/genetics , Neoplasms/metabolism , Signal Transduction/genetics , Stress, Physiological/metabolism , Transcription Factor CHOP/metabolism , eIF-2 Kinase/genetics
15.
Trends Biochem Sci ; 30(8): 426-9, 2005 Aug.
Article in English | MEDLINE | ID: mdl-15996866

ABSTRACT

Tumor hypoxia activates hypoxia-inducible factor-1 (HIF-1) and induces the accumulation of the tumor suppressor p53. HIF-1 signaling stimulates angiogenesis and mediates cellular adaptation to hypoxia, whereas p53 promotes hypoxia-induced apoptosis. A recent article provides in vitro biophysical evidence supporting a direct interaction between p53 and the oxygen-dependent degradation domain of the HIF-1alpha subunit. The article identifies potential structural parameters required for this interaction and suggests an alternative mechanism by which p53 might impact tumor response to therapy.


Subject(s)
Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Oxygen/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Cell Hypoxia/physiology , Humans , Protein Binding , Signal Transduction
16.
Semin Cell Dev Biol ; 16(4-5): 487-501, 2005.
Article in English | MEDLINE | ID: mdl-15896987

ABSTRACT

Hypoxia is a common feature of most solid tumors which negatively impacts their treatment response. This is due in part to the biological changes that result from a coordinated cellular response to hypoxia. A large part of this response is driven by a transcriptional program initiated via stabilization of HIF, promoting both angiogenesis and cell survival. However, hypoxia also results in a rapid inhibition of protein synthesis which occurs through the repression of the initiation step of mRNA translation. This inhibition is fully reversible and occurs in all cell lines tested to date. Inhibition of translation is mediated by two distinct mechanisms during hypoxia. The first is through phosphorylation and inhibition of an essential eukaryotic initiation factor, eIF2alpha. Phosphorylation of this factor occurs through activation of the PERK kinase as part of a coordinated ER stress response program known as the UPR. Activation of this program promotes cell survival during hypoxia and facilitates tumor growth. Translation during hypoxia can also be inhibited through the inactivation of a second eukaryotic initiation complex, eIF4F. At least part of this inhibition is mediated through a REDD1 and TSC1/TSC2 dependent inhibition of the mTOR kinase. Inhibition of mRNA translation is hypothesized to affect the cellular tolerance to hypoxia in part by promoting energy homeostasis. However, regulation of translation also results in a specific increase in the synthesis of a subset of hypoxia induced proteins. Consequently, both arms of translational control during hypoxia influence hypoxia induced gene expression and the hypoxic phenotype.


Subject(s)
Gene Expression Regulation/physiology , Hypoxia/metabolism , Oxidative Stress/physiology , Protein Biosynthesis/genetics , Protein Biosynthesis/physiology , RNA, Messenger/metabolism , Animals , Humans , Hypoxia/enzymology , Hypoxia/genetics , Oxidative Stress/genetics , RNA, Messenger/chemistry
SELECTION OF CITATIONS
SEARCH DETAIL
...