Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 17 de 17
Filter
1.
Mol Cancer Ther ; 23(4): 577-588, 2024 Apr 02.
Article in English | MEDLINE | ID: mdl-38359816

ABSTRACT

Treatments involving radiation and chemotherapy alone or in combination have improved patient survival and quality of life. However, cancers frequently evade these therapies due to adaptation and tumor evolution. Given the complexity of predicting response based solely on the initial genetic profile of a patient, a predetermined treatment course may miss critical adaptation that can cause resistance or induce new targets for drug and immunotherapy. To address the timescale for these evasive mechanisms, using a mouse xenograft tumor model, we investigated the rapidity of gene expression (mRNA), molecular pathway, and phosphoproteome changes after radiation, an HSP90 inhibitor, or combination. Animals received radiation, drug, or combination treatment for 1 or 2 weeks and were then euthanized along with a time-matched untreated group for comparison. Changes in gene expression occur as early as 1 week after treatment initiation. Apoptosis and cell death pathways were activated in irradiated tumor samples. For the HSP90 inhibitor and combination treatment at weeks 1 and 2 compared with Control Day 1, gene-expression changes induced inhibition of pathways including invasion of cells, vasculogenesis, and viral infection among others. The combination group included both drug-alone and radiation-alone changes. Our data demonstrate the rapidity of gene expression and functional pathway changes in the evolving tumor as it responds to treatment. Discovering these phenotypic adaptations may help elucidate the challenges in using sustained treatment regimens and could also define evolving targets for therapeutic efficacy.


Subject(s)
Antineoplastic Agents , Neoplasms , Animals , Humans , Heterografts , Multiomics , Quality of Life , Antineoplastic Agents/pharmacology , Neoplasms/drug therapy , Neoplasms/genetics , Neoplasms/radiotherapy , HSP90 Heat-Shock Proteins , Cell Line, Tumor , Xenograft Model Antitumor Assays
2.
Int J Mol Sci ; 22(9)2021 May 03.
Article in English | MEDLINE | ID: mdl-34063570

ABSTRACT

Understanding the global metabolic changes during the senescence of tumor cells can have implications for developing effective anti-cancer treatment strategies. Ionizing radiation (IR) was used to induce senescence in a human colon cancer cell line HCT-116 to examine secretome and metabolome profiles. Control proliferating and senescent cancer cells (SCC) exhibited distinct morphological differences and expression of senescent markers. Enhanced secretion of pro-inflammatory chemokines and IL-1, anti-inflammatory IL-27, and TGF-ß1 was observed in SCC. Significantly reduced levels of VEGF-A indicated anti-angiogenic activities of SCC. Elevated levels of tissue inhibitors of matrix metalloproteinases from SCC support the maintenance of the extracellular matrix. Adenylate and guanylate energy charge levels and redox components NAD and NADP and glutathione were maintained at near optimal levels indicating the viability of SCC. Significant accumulation of pyruvate, lactate, and suppression of the TCA cycle in SCC indicated aerobic glycolysis as the predominant energy source for SCC. Levels of several key amino acids decreased significantly, suggesting augmented utilization for protein synthesis and for use as intermediates for energy metabolism in SCC. These observations may provide a better understanding of cellular senescence basic mechanisms in tumor tissues and provide opportunities to improve cancer treatment.


Subject(s)
Cellular Senescence/genetics , Colonic Neoplasms/genetics , Metabolic Networks and Pathways/genetics , Metabolome/genetics , Cellular Senescence/radiation effects , Colonic Neoplasms/metabolism , Colonic Neoplasms/pathology , HCT116 Cells , Humans , Interleukin-1/genetics , Interleukin-27/genetics , Metabolic Networks and Pathways/radiation effects , Metabolome/radiation effects , Radiation, Ionizing , Transforming Growth Factor beta1/genetics , Vascular Endothelial Growth Factor A/genetics
3.
Int J Radiat Oncol Biol Phys ; 110(5): 1295-1305, 2021 08 01.
Article in English | MEDLINE | ID: mdl-33838214

ABSTRACT

PURPOSE: Recent preclinical studies suggest combining the HSP90 inhibitor AT13387 (Onalespib) with radiation (IR) against colon cancer and head and neck squamous cell carcinoma (HNSCC). These studies emphasized that AT13387 downregulates HSP90 client proteins involved in oncogenic signaling and DNA repair mechanisms as major drivers of enhanced radiosensitivity. Given the large array of client proteins HSP90 directs, we hypothesized that other key proteins or signaling pathways may be inhibited by AT13387 and contribute to enhanced radiosensitivity. Metabolomic analysis of HSP90 inhibition by AT13387 was conducted to identify metabolic biomarkers of radiosensitization and whether modulations of key proteins were involved in IR-induced tumor vasculogenesis, a process involved in tumor recurrence. METHODS AND MATERIALS: HNSCC and non-small cell lung cancer cell lines were used to evaluate the AT13387 radiosensitization effect in vitro and in vivo. Flow cytometry, immunofluorescence, and immunoblot analysis were used to evaluate cell cycle changes and HSP90 client protein's role in DNA damage repair. Metabolic analysis was performed using liquid chromatography-Mass spectrometry. Immunohistochemical examination of resected tumors post-AT13387 and IR treatment were conducted to identify biomarkers of IR-induced tumor vasculogenesis. RESULTS: In agreement with recent studies, AT13387 treatment combined with IR resulted in a G2/M cell cycle arrest and inhibited DNA repair. Metabolomic profiling indicated a decrease in key metabolites in glycolysis and tricarboxylic acid cycle by AT13387, a reduction in Adenosine 5'-triphosphate levels, and rate-limiting metabolites in nucleotide metabolism, namely phosphoribosyl diphosphate and aspartate. HNSCC xenografts treated with the combination exhibited increased tumor regrowth delay, decreased tumor infiltration of CD45 and CD11b+ bone marrow-derived cells, and inhibition of HIF-1 and SDF-1 expression, thereby inhibiting IR-induced vasculogenesis. CONCLUSIONS: AT13387 treatment resulted in pharmacologic inhibition of cancer cell metabolism that was linked to DNA damage repair. AT13387 combined with IR inhibited IR-induced vasculogenesis, a process involved in tumor recurrence postradiotherapy. Combining AT13387 with IR warrants consideration of clinical trial assessment.


Subject(s)
Benzamides/pharmacology , DNA Repair , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Head and Neck Neoplasms/radiotherapy , Isoindoles/pharmacology , Radiation Tolerance/drug effects , Squamous Cell Carcinoma of Head and Neck/radiotherapy , Animals , Aspartic Acid/pharmacology , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Cycle/drug effects , Cell Cycle/radiation effects , Cell Line, Tumor , Colonic Neoplasms/radiotherapy , DNA Damage , DNA Repair/drug effects , DNA Repair/radiation effects , Down-Regulation , G2 Phase Cell Cycle Checkpoints/drug effects , G2 Phase Cell Cycle Checkpoints/radiation effects , HSP90 Heat-Shock Proteins/metabolism , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/metabolism , Humans , Lung Neoplasms/radiotherapy , M Phase Cell Cycle Checkpoints/drug effects , M Phase Cell Cycle Checkpoints/radiation effects , Metabolomics , Mice , Mice, Nude , Neoplasm Recurrence, Local , Neovascularization, Pathologic/etiology , Neovascularization, Pathologic/prevention & control , Nucleotides/biosynthesis , Nucleotides/metabolism , Radiation Tolerance/genetics , Squamous Cell Carcinoma of Head and Neck/genetics , Squamous Cell Carcinoma of Head and Neck/metabolism , Xenograft Model Antitumor Assays
4.
Food Addit Contam Part B Surveill ; 13(4): 284-291, 2020 Dec.
Article in English | MEDLINE | ID: mdl-32552602

ABSTRACT

Food adulteration has a direct impact on public health, religious faith, fair-trades, and wildlife. In the present study, a reliable and sensitive assay has been developed for verifying meat adulteration in food chain. The multiplex PCR system was optimised for identification of chicken, cow/buffalo, sheep/goat, horse/donkey, pork, and dog DNAs in a single reaction mixture simultaneously. The primers were designed using 12 S rRNA gene sequences with fragment size in the range of 113 bp to 800 bp, which can be easily visualised on agarose gel electrophoresis making the technique economical. After validation of accuracy, specificity, and sensitivity, commercially available meat products (n = 190) were screened, comprising both raw and cooked meat samples. The results demonstrated a high rate of adulteration (54.5%) in meat products. The technique developed here can be easily used for screening of different meat products for export and import purposes as well as for food inspection and livestock diagnostic laboratories.


Subject(s)
Food Contamination/analysis , Meat/analysis , Meat/classification , Multiplex Polymerase Chain Reaction/methods , Animals , Buffaloes/genetics , Cattle/genetics , Chickens/genetics , DNA/analysis , Dogs/genetics , Equidae/genetics , Goats/genetics , Horses/genetics , RNA, Ribosomal/genetics , Sensitivity and Specificity , Sheep/genetics , Swine/genetics
5.
Oncotarget ; 10(12): 1230-1232, 2019 Feb 08.
Article in English | MEDLINE | ID: mdl-30815224
6.
Semin Radiat Oncol ; 29(1): 81-89, 2019 01.
Article in English | MEDLINE | ID: mdl-30573188

ABSTRACT

The aberrant vasculature in the tumor microenvironment creates hypoxic zones, poor perfusion, and high interstitial fluid pressure. Also, the tumor cell metabolic phenotype utilizes the aerobic glycolytic pathways for energy source and generation of cell mass. These physiologic and metabolic phenotypes in solid tumors are amenable for molecular imaging techniques to extract imaging biomarkers such as pO2 and enzyme kinetics reflecting glycolysis. The imaging biomarkers have value in diagnostic and prognostic purposes. Additionally, they can be used to guide choices for tailored treatment regimens. Electron paramagnetic resonance imaging for pO2 imaging and 13C magnetic resonance imaging with hyperpolarized 13C probes such as 13C-labeled pyruvate have shown significant potential in characterizing the tumor microenvironment physiologically and metabolically.


Subject(s)
Molecular Imaging/methods , Neoplasms/diagnostic imaging , Neoplasms/metabolism , Neoplasms/radiotherapy , Tumor Microenvironment/radiation effects , Animals , Biomarkers, Tumor/metabolism , Glycolysis , Humans
7.
Cancer Res ; 78(24): 6828-6837, 2018 12 15.
Article in English | MEDLINE | ID: mdl-30301838

ABSTRACT

: Hypoxic zones in solid tumors contribute to radioresistance, and pharmacologic agents that increase tumor oxygenation prior to radiation, including antiangiogenic drugs, can enhance treatment response to radiotherapy. Although such strategies have been applied, imaging assessments of tumor oxygenation to identify an optimum time window for radiotherapy have not been fully explored. In this study, we investigated the effects of α-sulfoquinovosylacyl-1,3-propanediol (SQAP or CG-0321; a synthetic derivative of an antiangiogenic agent) on the tumor microenvironment in terms of oxygen partial pressure (pO2), oxyhemoglobin saturation (sO2), blood perfusion, and microvessel density using electron paramagnetic resonance imaging, photoacoustic imaging, dynamic contrast-enhanced MRI with Gd-DTPA injection, and T2*-weighted imaging with ultrasmall superparamagnetic iron oxide (USPIO) contrast. SCCVII and A549 tumors were grown by injecting tumor cells into the hind legs of mice. Five days of daily radiation (2 Gy) combined with intravenous injection of SQAP (2 mg/kg) 30 minutes prior to irradiation significantly delayed growth of tumor xenografts. Three days of daily treatment improved tumor oxygenation and decreased tumor microvascular density on T2*-weighted images with USPIO, suggesting vascular normalization. Acute effects of SQAP on tumor oxygenation were examined by pO2, sO2, and Gd-DTPA contrast-enhanced imaging. SQAP treatment improved perfusion and tumor pO2 (ΔpO2: 3.1 ± 1.0 mmHg) and was accompanied by decreased sO2 (20%-30% decrease) in SCCVII implants 20-30 minutes after SQAP administration. These results provide evidence that SQAP transiently enhanced tumor oxygenation by facilitating oxygen dissociation from oxyhemoglobin and improving tumor perfusion. Therefore, SQAP-mediated sensitization to radiation in vivo can be attributed to increased tumor oxygenation. SIGNIFICANCE: A multimodal molecular imaging study evaluates pharmacological alteration of the tumor microenvironment to improve radiation response.


Subject(s)
Molecular Imaging/methods , Multimodal Imaging/methods , Neoplasms/drug therapy , Neoplasms/radiotherapy , Tumor Microenvironment , A549 Cells , Acoustics , Angiogenesis Inhibitors/pharmacology , Animals , Electron Spin Resonance Spectroscopy , Gadolinium/chemistry , Gadolinium DTPA/chemistry , Glycolipids/pharmacology , Human Umbilical Vein Endothelial Cells , Humans , Hypoxia , Magnetic Resonance Imaging , Mice , Mice, Inbred C3H , Microcirculation , Neoplasm Transplantation , Neoplasms/metabolism , Oxygen/chemistry , Oxygen/metabolism , Photochemistry , Radiation Tolerance , Radiotherapy
8.
Clin Cancer Res ; 24(16): 3994-4005, 2018 08 15.
Article in English | MEDLINE | ID: mdl-29716919

ABSTRACT

Purpose: To characterize the ionizing radiation (IR) enhancing effects and underlying mechanisms of the CDK4/6 inhibitor abemaciclib in non-small cell lung cancer (NSCLC) cells in vitro and in vivoExperimental Design: IR enhancement by abemaciclib in a variety of NSCLC cell lines was assessed by in vitro clonogenic assay, flow cytometry, and target inhibition verified by immunoblotting. IR-induced DNA damage repair was evaluated by γH2AX analysis. Global metabolic alterations by abemaciclib and IR combination were evaluated by LC/MS mass spectrometry and YSI bioanalyzer. Effects of abemaciclib and IR combination in vivo were studied by xenograft tumor regrowth delay, xenograft lysate immunoblotting, and tissue section immunohistochemistry.Results: Abemaciclib enhanced the radiosensitivity of NSCLC cells independent of RAS or EGFR status. Enhancement of radiosensitivity was lost in cell lines deficient for functional p53 and RB protein. After IR, abemaciclib treatment inhibited DNA damage repair as measured by γH2AX. Mechanistically, abemaciclib inhibited RB phosphorylation, leading to cell-cycle arrest. It also inhibited mTOR signaling and reduced intracellular amino acid pools, causing nutrient stress. In vivo, abemaciclib, when administered in an adjuvant setting for the second week after fractionated IR, further inhibited vasculogenesis and tumor regrowth, with sustained inhibition of RB/E2F activity, mTOR pathway, and HIF-1 expression. In summary, our study signifies inhibiting the CDK4/6 pathway by abemaciclib in combination with IR as a promising therapeutic strategy to treat NSCLC.Conclusions: Abemaciclib in combination with IR enhances NSCLC radiosensitivity in preclinical models, potentially providing a novel biomarker-driven combination therapeutic strategy for patients with NSCLC. Clin Cancer Res; 24(16); 3994-4005. ©2018 AACR.


Subject(s)
Aminopyridines/pharmacology , Benzimidazoles/pharmacology , Carcinoma, Non-Small-Cell Lung/drug therapy , Neovascularization, Pathologic/drug therapy , Protein Kinase Inhibitors/pharmacology , A549 Cells , Animals , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/pathology , Carcinoma, Non-Small-Cell Lung/radiotherapy , Cell Proliferation/drug effects , Cell Proliferation/radiation effects , Combined Modality Therapy , Cyclin-Dependent Kinase 4/antagonists & inhibitors , Cyclin-Dependent Kinase 4/genetics , Cyclin-Dependent Kinase 6/antagonists & inhibitors , Cyclin-Dependent Kinase 6/genetics , DNA Damage/drug effects , DNA Damage/radiation effects , DNA Repair/drug effects , DNA Repair/radiation effects , ErbB Receptors/genetics , Heterografts , Histones/genetics , Humans , Mice , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Neovascularization, Pathologic/radiotherapy , Radiation Tolerance/drug effects , Radiation, Ionizing , Xenograft Model Antitumor Assays
9.
FASEB J ; 32(8): 4145-4157, 2018 08.
Article in English | MEDLINE | ID: mdl-29494264

ABSTRACT

Deregulation of mitochondrial morphogenesis, a dynamic equilibrium between mitochondrial fusion and fission processes, is now evolving as a key metabolic event that fuels tumor growth and therapy resistance. However, fundamental knowledge underpinning how cancer cells reprogram mitochondrial morphogenesis remains incomplete. Here, we report that cystathionine ß-synthase (CBS) reprograms mitochondrial morphogenesis in ovarian cancer (OvCa) cells by selectively regulating the stability of mitofusin 2 (MFN2). Clinically, high expression of both CBS and MFN2 implicates poor overall survival of OvCa patients, and a significant association between CBS and MFN2 expression exists in individual patients in the same data set. The silencing of CBS by small interfering RNA or inhibition of its catalytic activity by a small molecule inhibitor creates oxidative stress that activates JNK. Activated JNK phosphorylates MFN2 to recruit homologous to the E6-AP carboxyl terminus' domain-containing ubiquitin E3 ligase for its degradation via the ubiquitin-proteasome system. Supplementation with hydrogen sulfide or glutathione (the catalytic products of CBS enzymatic activity), anti-oxidants, or a JNK inhibitor restores MFN2 expression. In CBS-silenced orthotopic xenograft tumor tissues, MFN2 but not MFN1 is selectively downregulated. In summary, this report reveals a role for deregulated mitochondrial morphogenesis in OvCa, suggests one of the mechanisms for this deregulation, and provides a way to correct it through modulation of the metabolic enzyme CBS.-Chakraborty, P. K., Murphy, B., Mustafi, S. B., Dey, A., Xiong, X., Rao, G., Naz, S., Zhang, M., Yang, D., Dhanasekaran, D. N., Bhattacharya, R., Mukherjee, P. Cystathionine ß-synthase regulates mitochondrial morphogenesis in ovarian cancer.


Subject(s)
Cystathionine beta-Synthase/metabolism , Mitochondria/metabolism , Mitochondrial Proteins/metabolism , Ovarian Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Down-Regulation/physiology , Female , Humans , MAP Kinase Signaling System/physiology , Oxidative Stress/physiology
10.
Radiat Res ; 189(5): 490-496, 2018 05.
Article in English | MEDLINE | ID: mdl-29528769

ABSTRACT

Amifostine is a potent antioxidant that protects against ionizing radiation effects. In this study, we evaluated the effect of Amifostine administered before total-body irradiation (TBI), at a drug dose that protects against TBI lethality, for potential protection against radiation-induced late effects such as a shortened lifespan and cancer. Three groups of mice were studied: 0 Gy control; 10.8 Gy TBI with Amifostine pretreatment; and 5.4 Gy TBI alone. Animals were monitored for their entire lifespan. The median survival times for mice receiving 0, 5.4 or 10.8 Gy TBI were 706, 460 and 491 days, respectively. Median survival of both irradiated groups was significantly shorter compared to nonirradiated mice ( P < 0.0001). Cancer incidence (hematopoietic and solid tumors) was similar between the irradiated groups and was significantly greater than for the 0 Gy controls. The ratio of hematopoietic-to-solid tumors differed among the groups, with the 5.4 Gy group having a higher incidence of hematopoietic neoplasms compared to the 10.8 Gy/Amifostine group (1.8-fold). Solid tumor incidence was greater in the 10.8 Gy/Amifostine group (1.6-fold). There are few mouse lifespan studies for agents that protect against radiation-induced lethality. Mice treated with 10.8 Gy/Amifostine yielded a lower incidence of hematopoietic neoplasms and higher incidence of solid neoplasms. In conclusion, mice protected from lethal TBI have a shortened lifespan, due in large part to cancer induction after exposure compared to nonexposed controls. Amifostine treatment did protect against radiation-induced hematopoietic tumors, while protection against solid neoplasms was significant but incomplete.


Subject(s)
Amifostine/pharmacology , Neoplasms, Radiation-Induced/prevention & control , Radiation-Protective Agents/pharmacology , Whole-Body Irradiation/adverse effects , Animals , Carcinogenesis/drug effects , Carcinogenesis/radiation effects , Dose-Response Relationship, Radiation , Female , Mice , Neoplasms, Radiation-Induced/etiology
11.
Antioxid Redox Signal ; 28(2): 131-140, 2018 Jan 10.
Article in English | MEDLINE | ID: mdl-28741367

ABSTRACT

AIMS: Evofosfamide (TH-302) is a hypoxia-activated prodrug (HAP) that releases the DNA-damaging bromo-isophosphoramide mustard (Br-IPM) moiety selectively under hypoxic conditions. Since solid tumors are known to have hypoxic regions, HAPs in combination with chemotherapy or radiotherapy (XRT) will be beneficial. We tested the oxygen dependence of release kinetics of Br-IPM using electron paramagnetic resonance (EPR) with spin trapping by monitoring redox cycling of the nitroimidazole moiety of TH-302, and oxygen dependence of TH-302 on in vitro cytotoxicity at different levels of hypoxia was also examined. Two tumor implants (SCCVII and HT29) in mice were studied. RESULTS: TH-302 fragmentation to release Br-IPM was noticed at oxygen levels <76 mmHg, which increased with higher levels of hypoxia. Enhanced cellular cytotoxicity was also observed at oxygen levels <76 mmHg. In vivo pO2 imaging in the two tumor implants showed that the SCCVII tumor implant had higher level of hypoxia compared with the HT29 xenograft. TH-302 as a monotherapy in vivo showed modest effects in SCCVII implants and minimal effects in HT29 xenografts, whereas TH-302 in combination with ionizing radiation showed significant benefit in both tumor models. INNOVATION: We examined the kinetics of redox cycling versus fragmentation of TH-302. The combination of oxygen-dependent XRT with TH-302 is effective even in tumors with significant hypoxia. CONCLUSIONS: Imaging studies identifying the magnitude of hypoxia in tumors indicated that the responsiveness to TH-302 and the antitumor effect of TH-302 were enhanced by combining with XRT in both the TH-302-sensitive SCCVII tumor and -resistant HT29 tumor. Antioxid. Redox Signal. 28, 131-140.


Subject(s)
Hypoxia/metabolism , Nitroimidazoles/pharmacology , Phosphoramide Mustards/pharmacology , Prodrugs , Radiotherapy , Animals , Cell Hypoxia/drug effects , Cell Hypoxia/radiation effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , Disease Models, Animal , Humans , Hypoxia/drug therapy , Hypoxia/radiotherapy , Mice , Oxidation-Reduction/drug effects , Oxidation-Reduction/radiation effects , Tumor Burden/drug effects , Tumor Burden/radiation effects , Xenograft Model Antitumor Assays
12.
Biochim Biophys Acta ; 1859(8): 983-93, 2016 08.
Article in English | MEDLINE | ID: mdl-27295567

ABSTRACT

Chemotherapy-induced emergence of drug resistant cells is frequently observed and is exemplified by the expression of family of drug resistance proteins including, multidrug resistance protein 1 (MDR1). However, a concise mechanism for chemotherapy-induced MDR1 expression is unclear. Mechanistically, mutational selection, epigenetic alteration, activation of the Wnt pathway or impaired p53 function have been implicated. The present study describes that the surviving fraction of cisplatin resistant cells co- upregulate MDR1, BMI1 and acetyl transferase activity of TIP60. Using complementary gain and loss of function approaches, we demonstrate that the expression of MDR1 is positively regulated by BMI1, a stem-cell factor classically known as a transcriptional repressor. Our study establishes a functional interaction between TIP60 and BMI-1 resulting in upregulation of MDR1 expression. Chromatin immunoprecipitation (ChIP) assays further establish that the proximal MDR1 promoter responds to cisplatin in a BMI1 dependent manner. BMI1 interacts with a cluster of E-box elements on the MDR1 promoter and recruits TIP60 resulting in acetylation of histone H2A and H3. Collectively, our data establish a hitherto unknown liaison among MDR1, BMI1 and TIP60 and provide mechanistic insights into cisplatin-induced MDR1 expression resulting in acquired cross-resistance against paclitaxel, doxorubicin and likely other drugs. In conclusion, our results advocate utilizing anti-BMI1 strategies to alleviate acquired resistance to chemotherapy.


Subject(s)
Antineoplastic Agents/pharmacology , Drug Resistance, Multiple/drug effects , Drug Resistance, Neoplasm/drug effects , Gene Expression Regulation, Neoplastic , Histone Acetyltransferases/genetics , Polycomb Repressive Complex 1/genetics , ATP Binding Cassette Transporter, Subfamily B/agonists , ATP Binding Cassette Transporter, Subfamily B/genetics , ATP Binding Cassette Transporter, Subfamily B/metabolism , Acetylation/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Cisplatin/pharmacology , Doxorubicin/pharmacology , Drug Resistance, Multiple/genetics , Drug Resistance, Neoplasm/genetics , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Histone Acetyltransferases/metabolism , Histones/genetics , Histones/metabolism , Humans , Lysine Acetyltransferase 5 , Paclitaxel/pharmacology , Polycomb Repressive Complex 1/agonists , Polycomb Repressive Complex 1/metabolism , Promoter Regions, Genetic , Protein Binding , Signal Transduction
13.
Genetics ; 200(1): 149-65, 2015 May.
Article in English | MEDLINE | ID: mdl-25747460

ABSTRACT

Rapid and high wing-beat frequencies achieved during insect flight are powered by the indirect flight muscles, the largest group of muscles present in the thorax. Any anomaly during the assembly and/or structural impairment of the indirect flight muscles gives rise to a flightless phenotype. Multiple mutagenesis screens in Drosophila melanogaster for defective flight behavior have led to the isolation and characterization of mutations that have been instrumental in the identification of many proteins and residues that are important for muscle assembly, function, and disease. In this article, we present a molecular-genetic characterization of a flightless mutation, flightless-H (fliH), originally designated as heldup-a (hdp-a). We show that fliH is a cis-regulatory mutation of the wings up A (wupA) gene, which codes for the troponin-I protein, one of the troponin complex proteins, involved in regulation of muscle contraction. The mutation leads to reduced levels of troponin-I transcript and protein. In addition to this, there is also coordinated reduction in transcript and protein levels of other structural protein isoforms that are part of the troponin complex. The altered transcript and protein stoichiometry ultimately culminates in unregulated acto-myosin interactions and a hypercontraction muscle phenotype. Our results shed new insights into the importance of maintaining the stoichiometry of structural proteins during muscle assembly for proper function with implications for the identification of mutations and disease phenotypes in other species, including humans.


Subject(s)
Drosophila Proteins/genetics , Drosophila melanogaster/genetics , Mutation , Protein Multimerization , Regulatory Sequences, Nucleic Acid , Sarcomeres/metabolism , Troponin I/genetics , Animals , Drosophila Proteins/metabolism , Drosophila melanogaster/metabolism , Muscle Contraction , Protein Isoforms/genetics , Protein Isoforms/metabolism , RNA, Messenger/genetics , RNA, Messenger/metabolism , Sarcomeres/physiology , Troponin I/metabolism
14.
Carcinogenesis ; 35(1): 14-23, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23996929

ABSTRACT

S100 family of calcium-binding proteins is commonly upregulated in a variety of tumor types and is often associated with tumor progression. Among several S100 members, altered expression of S100A2 is a potential diagnostic and prognostic marker in cancer. Several reports suggest a role for S100A2 in metastasis. Earlier, our studies established regulation of S100A2 by transforming growth factor-ß (TGF-ß) and its involvement in TGF-ß-mediated cancer cell invasion and migration. However, the molecular mechanisms of S100A2 protumorigenic actions remain unexplored. In the present study, we demonstrate that overexpression of S100A2 in A549 lung cancer cells induced epithelial-mesenchymal transition (EMT) followed by increased invasion, loose colony morphology in soft agar and enhanced Akt phosphorylation (Ser-473). Furthermore, overexpression of S100A2 led to increased tumor growth in immunocompromised mice. In agreement, immunohistochemical examination of resected xenograft tumors established inverse correlation between S100A2 and E-cadherin expression together with activated Akt signaling. Interestingly, our study demonstrates a strong dependence of S100A2 and Smad3 in TGF-ß-induced Hep3B cell EMT and invasion. Most importantly, we demonstrate that these effects of S100A2 are manifested through functional interaction with Smad3, which is enhanced in the presence of high calcium and TGF-ß. S100A2 stabilizes Smad3 and binds to its C-terminal MH2 domain. Additionally, loss of S100A2 attenuates the transcription of TGF-ß/Smad3 target genes involved in tumor promotion, such as PA1-1 and vimentin. Collectively, our findings present the first mechanistic details of S100A2 protumorigenic actions and its involvement in TGF-ß-mediated cancer cell invasion and EMT.


Subject(s)
Chemotactic Factors/metabolism , Lung Neoplasms/metabolism , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins c-akt/metabolism , S100 Proteins/metabolism , Smad3 Protein/metabolism , Animals , Calcium/metabolism , Calcium/pharmacology , Cell Line, Tumor , Cell Proliferation , Chemotactic Factors/genetics , Epithelial-Mesenchymal Transition/drug effects , Female , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/pathology , Mice, Nude , Protein Structure, Tertiary , S100 Proteins/genetics , Signal Transduction , Smad3 Protein/genetics , Transforming Growth Factor beta/metabolism , Transforming Growth Factor beta/pharmacology , Xenograft Model Antitumor Assays
15.
Biomaterials ; 35(4): 1334-46, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24211075

ABSTRACT

The p53 protein mediated anti-tumor strategy is limited due to the lack of suitable delivery agent with insignificant immunogenic response, serum compatibility, and early and easy detection of the transfected cell population. To overcome these problems, we generated a p53-EGFP-C3 fusion construct which expressed easily detectable green fluorescence protein (GFP) and allowed an estimation of p53 mediated anti-tumor activity. A mixture of cationic cholesterol gemini (Chol-5L) with natural lipid, DOPE (molar ratio 1:4), acronymed as Chol-5LD, formed a nano-liposome as characterized by various physical methods. The prepared clone was evaluated for the expression of GFP and functional p53 in HeLa and two additional cell lines with varied p53 status namely, H1299 (p53(-/-)) and HEK293T (p53(+/+)). Transfected cells were screened using RT-PCR, Western blotting, FACS analysis, MTT, Trypan blue assay and visualized under a fluorescence microscope. The p53-EGFP-C3 fusion protein induced apoptosis in cancer cells as evident from DNA fragmentation, cell cycle analysis, Annexin-V staining and PARP cleavage assays. The transfection and apoptosis induction efficiency of Chol-5LD was significantly higher than commercial reagents Lipofectamine2000 and Effectene irrespective of the cell lines examined. Further it significantly decreases the xenograft tumor volume in nude mice tumors via apoptosis as observed in H&E staining.


Subject(s)
Cholesterol/chemistry , Gene Transfer Techniques , Green Fluorescent Proteins/genetics , Nanostructures/chemistry , Neoplasms/therapy , Plasmids/therapeutic use , Tumor Suppressor Protein p53/genetics , Animals , Apoptosis , Cations/chemistry , Cell Line, Tumor , Genetic Therapy , HEK293 Cells , HeLa Cells , Humans , Mice , Mice, Nude , Neoplasms/genetics , Neoplasms/pathology , Plasmids/genetics
16.
ACS Chem Biol ; 8(7): 1429-34, 2013 Jul 19.
Article in English | MEDLINE | ID: mdl-23621494

ABSTRACT

In the search for more efficacious and less toxic cancer drugs, the tumor suppressor p53 protein has long been a desirable therapeutic target. In the recent past, few independent studies have demonstrated that the antitumor activity of wild-type p53 can be restored in cancer cells harboring mutant form of p53 using small molecule activators. In this study, we describe a novel small molecule MPK-09, which is selective and highly potent against allele specific p53 mutations mainly, R175H, R249S, R273H, R273C, and E285K . Except E285K, all other mutations tested are among the six "hot spot" p53 mutations reported in majority of human cancer. Furthermore, our study conclusively demonstrates that the apoptotic activity of the small molecule MPK-09 against cancer cells harboring R273C and E285K mutations is due to restoration of the wild-type conformation to the corresponding mutant form of p53.


Subject(s)
Apoptosis/drug effects , Butyrates/chemistry , Butyrates/pharmacology , Ethers, Cyclic/chemistry , Ethers, Cyclic/pharmacology , Tumor Suppressor Protein p53/genetics , Blotting, Western , Cell Line, Tumor , Flow Cytometry , Fluorescent Antibody Technique , Gene Deletion , Humans , Inhibitory Concentration 50 , Lactones/chemistry , Lactones/pharmacology , Molecular Structure , Mutation , Styrenes/chemistry , Styrenes/pharmacology
17.
Biochem J ; 447(1): 81-91, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-22747445

ABSTRACT

S100A2, an EF hand calcium-binding protein, is a potential biomarker in several cancers and is also a TGF-ß (transforming growth factor-ß)-regulated gene in melanoma and lung cancer cells. However, the mechanism of S100A2 regulation by TGF-ß and its significance in cancer progression remains largely unknown. In the present study we report the mechanism of S100A2 regulation by TGF-ß and its possible role in TGF-ß-mediated tumour promotion. Characterization of the S100A2 promoter revealed an AP-1 (activator protein-1) element at positions -1161 to -1151 as being the most critical factor for the TGF-ß1 response. Chromatin immunoprecipitation and electrophoretic mobility-shift assays confirmed the functional binding of the AP-1 complex, predominantly JunB, to the S100A2 promoter in response to TGF-ß1 in HaCaT keratinocytes. JunB overexpression markedly stimulated the S100A2 promoter which was blocked by the dominant-negative JunB and MEK1 [MAPK (mitogen-activated protein kinase)/ERK (extracellular-signal-regulated kinase) kinase 1] inhibitor, PD98059. Intriguingly, despite the presence of a putative SMAD-binding element, S100A2 regulation by TGF-ß1 was found to be SMAD3 independent. Interestingly, p53 protein and TGF-ß1 show synergistic regulation of the S100A2 promoter. Finally, knockdown of S100A2 expression compromised TGF-ß1-induced cell migration and invasion of Hep3B cells. Together our findings highlight an important link between the TGF-ß1-induced MAPK and p53 signalling pathways in the regulation of S100A2 expression and pro-tumorigenic actions.


Subject(s)
Chemotactic Factors/genetics , MAP Kinase Signaling System/drug effects , Neoplasm Invasiveness/physiopathology , S100 Proteins/genetics , Transforming Growth Factor beta1/pharmacology , Cell Line , Cell Line, Tumor , Cell Movement/drug effects , Cell Movement/physiology , Chemotactic Factors/antagonists & inhibitors , Chemotactic Factors/physiology , Female , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Humans , Promoter Regions, Genetic , Proto-Oncogene Proteins c-jun/metabolism , RNA, Small Interfering/genetics , S100 Proteins/antagonists & inhibitors , S100 Proteins/physiology , Transcription Factor AP-1/metabolism , Tumor Suppressor Protein p53/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...