Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 56
Filter
1.
Int J Mol Sci ; 22(5)2021 Mar 02.
Article in English | MEDLINE | ID: mdl-33801310

ABSTRACT

BACKGROUND: The molecular regulation of increased MGMT expression in human brain tumors, the associated regulatory elements, and linkages of these to its epigenetic silencing are not understood. Because the heightened expression or non-expression of MGMT plays a pivotal role in glioma therapeutics, we applied bioinformatics and experimental tools to identify the regulatory elements in the MGMT and neighboring EBF3 gene loci. RESULTS: Extensive genome database analyses showed that the MGMT genomic space was rich in and harbored many undescribed RNA regulatory sequences and recognition motifs. We extended the MGMT's exon-1 promoter to 2019 bp to include five overlapping alternate promoters. Consensus sequences in the revised promoter for (a) the transcriptional factors CTCF, NRF1/NRF2, GAF, (b) the genetic switch MYC/MAX/MAD, and (c) two well-defined p53 response elements in MGMT intron-1, were identified. A putative protein-coding or non-coding RNA sequence was located in the extended 3' UTR of the MGMT transcript. Eleven non-coding RNA loci coding for miRNAs, antisense RNA, and lncRNAs were identified in the MGMT-EBF3 region and six of these showed validated potential for curtailing the expression of both MGMT and EBF3 genes. ChIP analysis verified the binding site in MGMT promoter for CTCF which regulates the genomic methylation and chromatin looping. CTCF depletion by a pool of specific siRNA and shRNAs led to a significant attenuation of MGMT expression in human GBM cell lines. Computational analysis of the ChIP sequence data in ENCODE showed the presence of NRF1 in the MGMT promoter and this occurred only in MGMT-proficient cell lines. Further, an enforced NRF2 expression markedly augmented the MGMT mRNA and protein levels in glioma cells. CONCLUSIONS: We provide the first evidence for several new regulatory components in the MGMT gene locus which predict complex transcriptional and posttranscriptional controls with potential for new therapeutic avenues.


Subject(s)
Biomarkers, Tumor/metabolism , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Epigenesis, Genetic , Gene Expression Regulation, Neoplastic , Glioma/metabolism , Regulatory Sequences, Nucleic Acid , Tumor Suppressor Proteins/metabolism , Biomarkers, Tumor/genetics , Brain Neoplasms/genetics , Brain Neoplasms/metabolism , Brain Neoplasms/pathology , CCCTC-Binding Factor/genetics , CCCTC-Binding Factor/metabolism , DNA Methylation , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Genomics , Glioma/genetics , Glioma/pathology , Humans , NF-E2-Related Factor 2/genetics , NF-E2-Related Factor 2/metabolism , Nuclear Respiratory Factor 1/genetics , Nuclear Respiratory Factor 1/metabolism , Promoter Regions, Genetic , RNA, Small Interfering/genetics , RNA, Untranslated/genetics , Tumor Suppressor Proteins/genetics
2.
Carcinogenesis ; 39(11): 1399-1410, 2018 12 13.
Article in English | MEDLINE | ID: mdl-30010803

ABSTRACT

The molecular basis of anticancer and apoptotic effects of R-goniothalamin (GON), a plant secondary metabolite was studied. We show that induction of oxidative stress and reactivation of mutant p53 underlie the strong cytotoxic effects of GON against the breast cancer cells. While GON was not toxic to the MCF10a breast epithelial cells, the SKBR3 breast cancer cells harboring an R175H mutant p53 were highly sensitive (IC50 = 7.3 µM). Flow cytometry and other pertinent assays showed that GON-induced abundant reactive oxygen species (ROS), glutathione depletion, protein glutathionylation and activation of apoptotic markers. GON was found to conjugate with glutathione both in vitro and in cells and the product was characterized by mass spectrometry. We hypothesized that the redox imbalance induced by GON may affect the structure of the R175H mutant p53 protein, and account for greater cytotoxicity. Using the SKBR3 breast cancer and p53-null H1299 lung cancer cells stably expressing the R175H p53 mutant protein, we demonstrated that GON triggers the appearance of a wild-type-like p53 protein by using conformation-specific antibodies, immunoprecipitation, DNA-binding assays and target gene expression. p53 restoration was associated with a G2/M arrest, senescence, reduced cell migration, invasion and increased cell death. GON elicited a highly synergistic cytotoxicity with cisplatin in SKBR3 cells. In SKBR3 xenografts developed in nude mice, there was a marked tumor growth delay by GON alone and GON + cisplatin combination. Our studies highlight the impact of tumor redox-stress generated by GON in activating the mutant p53 protein for greater antitumor efficacy.


Subject(s)
Antineoplastic Agents/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Glutathione/metabolism , Pyrones/pharmacology , Tumor Suppressor Protein p53/genetics , Animals , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Breast/cytology , Breast/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cisplatin/pharmacology , Female , G2 Phase Cell Cycle Checkpoints/drug effects , Humans , MCF-7 Cells , Mice , Mice, Nude , Neoplasm Invasiveness , Oxidation-Reduction/drug effects , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Xenograft Model Antitumor Assays
3.
Tumour Biol ; 39(6): 1010428317705517, 2017 Jun.
Article in English | MEDLINE | ID: mdl-28618969

ABSTRACT

Pancreatic cancer is one of the most aggressive and difficult to treat cancers. Experimental and clinical evidence suggests that high basal state autophagy in pancreatic tumors could induce resistance to chemotherapy. Recently, we have demonstrated that penfluridol suppresses pancreatic tumor growth by autophagy-mediated apoptosis both in vitro and in vivo; however, the mechanism of autophagy induction by penfluridol was not clear. Several studies have established that endoplasmic reticulum stress could lead to autophagy and inhibit tumor progression. In this study, we demonstrated that penfluridol induced endoplasmic reticulum stress in BxPC-3, AsPC-1, and Panc-1 pancreatic cancer cell lines as indicated by upregulation of endoplasmic reticulum stress markers such as binding protein (BIP), C/EBP homologous protein (CHOP) and inositol requiring 1α (IRE1α) after treatment with penfluridol in a concentration-dependent manner. Inhibiting endoplasmic reticulum stress by pretreatment with pharmacological inhibitors such as sodium phenylbutyrate and mithramycin or by silencing CHOP using CHOP small interfering RNA, blocked penfluridol-induced autophagy. These results clearly indicate that penfluridol-induced endoplasmic reticulum stress lead to autophagy in our model. Western blot analysis of subcutaneously implanted AsPC-1 and BxPC-3 tumors as well as orthotopically implanted Panc-1 tumors demonstrated upregulation of BIP, CHOP, and IRE1α expression in the tumor lysates from penfluridol-treated mice as compared to tumors from control mice. Altogether, our study establishes that penfluridol-induced endoplasmic reticulum stress leads to autophagy resulting in reduced pancreatic tumor growth. Our study opens a new therapeutic target for advanced chemotherapies against pancreatic cancer.


Subject(s)
Endoribonucleases/biosynthesis , Heat-Shock Proteins/genetics , Pancreatic Neoplasms/drug therapy , Penfluridol/administration & dosage , Protein Serine-Threonine Kinases/biosynthesis , Transcription Factor CHOP/biosynthesis , Animals , Apoptosis/drug effects , Autophagy/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Endoplasmic Reticulum/drug effects , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/drug effects , Endoribonucleases/genetics , Gene Expression Regulation, Neoplastic , Heat-Shock Proteins/biosynthesis , Humans , Mice , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Protein Serine-Threonine Kinases/genetics , RNA, Small Interfering/genetics , Transcription Factor CHOP/antagonists & inhibitors , Transcription Factor CHOP/genetics , Xenograft Model Antitumor Assays
4.
Bioorg Med Chem Lett ; 26(12): 2829-2833, 2016 06 15.
Article in English | MEDLINE | ID: mdl-27156773

ABSTRACT

Ethacrynic acid (EA), a known inhibitor of the neoplastic marker glutathione S-transferase P1 and other GSTs, exerts a weak antiproliferative activity against human cancer cells. The clinical use of EA (Edecrin) as an anticancer drug is limited by its potent loop diuretic activity. In this study, we developed a non-diuretic 2-amino-2-deoxy-d-glucose conjugated EA (EAG) to target tumors cells via the highly expressed glucose transporter 1 (GLUT1). Cell survival assays revealed that EAG had little effect on normal cells, but was cytotoxic 3 to 4.5-fold greater than EA. Mechanistically, the EAG induced selective cell death in cancer cells by inhibiting GSTP1 and generating abundant reactive oxygen species. Furthermore, EAG induced p21(cip1) expression and a G2/M cell cycle block irrespective of the p53 gene status in tumor cells. These data encourage the development of new EA analogs.


Subject(s)
Antineoplastic Agents/pharmacology , Enzyme Inhibitors/pharmacology , Ethacrynic Acid/pharmacology , Glucosamine/pharmacology , Antineoplastic Agents/chemical synthesis , Antineoplastic Agents/chemistry , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Drug Screening Assays, Antitumor , Enzyme Inhibitors/chemical synthesis , Enzyme Inhibitors/chemistry , Ethacrynic Acid/chemical synthesis , Glucosamine/analogs & derivatives , Glucosamine/chemistry , Glutathione S-Transferase pi/antagonists & inhibitors , Glutathione S-Transferase pi/metabolism , Humans , Molecular Structure , Structure-Activity Relationship
5.
Carcinogenesis ; 35(3): 692-702, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24193513

ABSTRACT

The alcohol aversion drug disulfiram (DSF) reacts and conjugates with the protein-bound nucleophilic cysteines and is known to elicit anticancer effects alone or improve the efficacy of many cancer drugs. We investigated the effects of DSF on human O(6)-methylguanine-DNA methyltransferase (MGMT), a DNA repair protein and chemotherapy target that removes the mutagenic O(6)-akyl groups from guanines, and thus confers resistance to alkylating agents in brain tumors. We used DSF, copper-chelated DSF or CuCl2-DSF combination and found that all treatments inhibited the MGMT activity in two brain tumor cell lines in a rapid and dose-dependent manner. The drug treatments resulted in the loss of MGMT protein from tumor cells through the ubiquitin-proteasome pathway. Evidence showed that Cys145, a reactive cysteine, critical for DNA repair was the sole site of DSF modification in the MGMT protein. DSF was a weaker inhibitor of MGMT, compared with the established O(6)-benzylguanine; nevertheless, the 24-36h suppression of MGMT activity in cell cultures vastly increased the alkylation-induced DNA interstrand cross-linking, G2/M cell cycle blockade, cytotoxicity and the levels of apoptotic markers. Normal mice treated with DSF showed significantly attenuated levels of MGMT activity and protein in the liver and brain tissues. In nude mice bearing T98 glioblastoma xenografts, there was a preferential inhibition of tumor MGMT. Our studies demonstrate a strong and direct inhibition of MGMT by DSF and support the repurposing of this brain penetrating drug for glioma therapy. The findings also imply an increased risk for alkylation damage in alcoholic patients taking DSF.


Subject(s)
Antineoplastic Agents, Alkylating/pharmacology , Brain Neoplasms/enzymology , Brain/drug effects , DNA Damage , Disulfiram/pharmacology , Enzyme Inhibitors/pharmacology , O(6)-Methylguanine-DNA Methyltransferase/antagonists & inhibitors , Animals , Brain/enzymology , Brain Neoplasms/pathology , Cell Line, Tumor , Humans , Mice
6.
Methods Mol Biol ; 2755: 63-74, 2024.
Article in English | MEDLINE | ID: mdl-38319569

ABSTRACT

Sensitive activity stains for enzymes selectively expressed in human cancers offer valuable tools for imaging with wide applications in experimental, diagnostic, and therapeutic settings. The scant expression of the antioxidant enzyme NQO1 in normal tissues and its great abundance in malignant counterparts due to the increased redox stress and hypoxia is one such example. Previously, we described a potent nontoxic probe that remains nonfluorescent but releases an intense fluorogenic compound after intracellular cleavage by NQO1 catalysis. This infrared probe with a 644 nm emission has excellent tissue penetrating ability and low background absorption. Described here are methods (fluorescence microscopy, flow cytometry, and in vivo animal imaging) to rapidly image NQO1 activity in hypoxic and non-hypoxic cancer cells and tumors developed in live mouse xenograft models. The specificity of the dye for NQO1 in all three procedures was verified, and the methods should be useful for both in vitro and in vivo studies.


Subject(s)
Neoplasms , Humans , Animals , Mice , Heterografts , Mice, Nude , Transplantation, Heterologous , Neoplasms/diagnostic imaging , Microscopy, Fluorescence , Disease Models, Animal , Hypoxia , NAD(P)H Dehydrogenase (Quinone)
7.
ACS Pharmacol Transl Sci ; 7(1): 120-136, 2024 Jan 12.
Article in English | MEDLINE | ID: mdl-38230276

ABSTRACT

Triple-negative breast cancer (TNBC) poses significant challenges due to its aggressive nature and limited treatment options. In this study, we investigated the impact of urea-based compounds on TNBC cells to uncover their mechanisms of action and therapeutic potential. Notably, polypharmacology urea analogues were found to work via p53-related pathways, and their cytotoxic effects were amplified by the modulation of oxidative phosphorylation pathways in the mitochondria of cancer cells. Specifically, compound 1 demonstrated an uncoupling effect on adenosine triphosphate (ATP) synthesis, leading to a time- and concentration-dependent shift toward glycolysis-based ATP production in MDA-MB-231 cells. At the same time, no significant changes in ATP synthesis were observed in noncancerous MCF10A cells. Moreover, the unique combination of mitochondrial- and p53-related effects leads to a higher cytotoxicity of urea analogues in cancer cells. Notably, the majority of tested clinical agents, but sorafenib, showed significantly higher toxicity in MCF10A cells. To test our hypothesis of sensitizing cancer cells to the treatment via modulation of mitochondrial health, we explored the combinatorial effects of urea-based analogues with established chemotherapeutic agents commonly used in TNBC treatment. Synergistic effects were evident in most tested combinations in TNBC cell lines, while noncancerous MCF10A cells exhibited higher resistance to these combination treatments. The combination of compound 1 with SN38 displayed nearly 60-fold selectivity toward TNBC cells over MCF10A cells. Encouragingly, combinations involving compound 1 restored the sensitivity of TNBC cells to cisplatin. In conclusion, our study provides valuable insights into the mechanisms of action of urea-based compounds in TNBC cells. The observed induction of mitochondrial membrane depolarization, inhibition of superoxide dismutase activity, disruption of ATP synthesis, and cell-line-specific responses contribute to their cytotoxic effects. Additionally, we demonstrated the synergistic potential of compound 1 to enhance the efficacy of existing TNBC treatments. However, the therapeutic potential and underlying molecular mechanisms of urea-based analogues in TNBC cell lines require further exploration.

8.
Carcinogenesis ; 34(5): 990-1000, 2013 May.
Article in English | MEDLINE | ID: mdl-23354308

ABSTRACT

The ionized cysteines present on the surfaces of many redox-sensitive proteins play functionally essential roles and are readily targeted by the reactive oxygen and reactive nitrogen species. Using disulfiram (DSF) and nitroaspirin (NCX4016) as the model compounds that mediate thiol-conjugating and nitrosylating reactions, respectively, we investigated the fate of p53, nuclear factor-kappaB (NF-κB) and other redox-responsive proteins following the exposure of human cancer cell lines to the drugs. Both drugs induced glutathionylation of bulk proteins in tumor cells and cell-free extracts. A prominent finding of this study was a time- and dose-dependent degradation of the redox-regulated proteins after brief treatments of tumor cells with DSF or NCX4016. DSF and copper-chelated DSF at concentrations of 50-200 µM induced the disappearance of wild-type p53, mutant p53, NF-κB subunit p50 and the ubiquitin-activating enzyme E1 (UBE1) in tumor cell lines. DSF also induced the glutathionylation of p53. The recombinant p53 protein modified by DSF was preferentially degraded by rabbit reticulocyte lysates. The proteasome inhibitor PS341 curtailed the DSF-induced degradation of p53 in HCT116 cells. Further, the NCX4016 induced a dose-dependent disappearance of the UBE1 and NF-κB p50 proteins in cell lines, besides a time-dependent degradation of aldehyde dehydrogenase in mouse liver after a single injection of 150 mg/kg. The loss of p53 and NF-kB proteins correlated with decreases in their specific binding to DNA. Our results demonstrate the hitherto unrecognized ability of the non-toxic thiolating and nitrosylating agents to degrade regulatory proteins and highlight the exploitable therapeutic benefits.


Subject(s)
NF-kappa B/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/pharmacology , Aspirin/analogs & derivatives , Aspirin/pharmacology , Cell Line, Tumor , DNA-Binding Proteins/genetics , DNA-Binding Proteins/metabolism , Disulfiram/pharmacology , HCT116 Cells , HT29 Cells , Humans , Liver/drug effects , Liver/metabolism , Male , Mice , NF-kappa B/genetics , Oxidation-Reduction/drug effects , Proteasome Endopeptidase Complex/drug effects , Proteasome Endopeptidase Complex/genetics , Proteasome Endopeptidase Complex/metabolism , Rabbits , Reticulocytes/drug effects , Reticulocytes/metabolism , Tumor Suppressor Protein p53/genetics , Ubiquitin-Activating Enzymes/genetics , Ubiquitin-Activating Enzymes/metabolism
9.
Mol Med ; 18: 913-29, 2012 Sep 07.
Article in English | MEDLINE | ID: mdl-22549111

ABSTRACT

Antiestrogen therapy resistance remains a huge stumbling block in the treatment of breast cancer. We have found significant elevation of O(6) methylguanine DNA methyl transferase (MGMT) expression in a small sample of consecutive patients who have failed tamoxifen treatment. Here, we show that tamoxifen resistance is accompanied by upregulation of MGMT. Further we show that administration of the MGMT inhibitor, O(6)-benzylguanine (BG), at nontoxic doses, leads to restoration of a favorable estrogen receptor alpha (ERα) phosphorylation phenotype (high p-ERα Ser167/low p-ERα Ser118), which has been reported to correlate with sensitivity to endocrine therapy and improved survival. We also show BG to be a dual inhibitor of MGMT and ERα. In tamoxifen-resistant breast cancer cells, BG alone or in combination with antiestrogen (tamoxifen [TAM]/ICI 182,780 [fulvestrant, Faslodex]) therapy enhances p53 upregulated modulator of apoptosis (PUMA) expression, cytochrome C release and poly (ADP-ribose) polymerase (PARP) cleavage, all indicative of apoptosis. In addition, BG increases the expression of p21(cip1/waf1). We also show that BG, alone or in combination therapy, curtails the growth of tamoxifen-resistant breast cancer in vitro and in vivo. In tamoxifen-resistant MCF7 breast cancer xenografts, BG alone or in combination treatment causes significant delay in tumor growth. Immunohistochemistry confirms that BG increases p21(cip1/waf1) and p-ERα Ser167 expression and inhibits MGMT, ERα, p-ERα Ser118 and ki-67 expression. Collectively, our results suggest that MGMT inhibition leads to growth inhibition of tamoxifen-resistant breast cancer in vitro and in vivo and resensitizes tamoxifen-resistant breast cancer cells to antiestrogen therapy. These findings suggest that MGMT inhibition may provide a novel therapeutic strategy for overcoming antiestrogen resistance.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/enzymology , DNA Modification Methylases/antagonists & inhibitors , DNA Repair Enzymes/antagonists & inhibitors , Estrogen Antagonists/therapeutic use , Estrogen Receptor alpha/metabolism , Tumor Suppressor Proteins/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Apoptosis/drug effects , Apoptosis/genetics , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Cycle Checkpoints/drug effects , Cell Cycle Checkpoints/genetics , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Enzyme Inhibitors/pharmacology , Enzyme Inhibitors/therapeutic use , Estrogen Antagonists/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Guanine/analogs & derivatives , Guanine/pharmacology , Guanine/therapeutic use , Humans , Immunohistochemistry , Mice , Mice, Nude , Phosphorylation/drug effects , Phosphoserine/metabolism , Promoter Regions, Genetic/genetics , Protein Binding/drug effects , Protein Binding/genetics , Tamoxifen/therapeutic use , Transcription, Genetic/drug effects , Tumor Suppressor Protein p53/metabolism , Tumor Suppressor Proteins/metabolism
10.
Genes (Basel) ; 12(6)2021 06 08.
Article in English | MEDLINE | ID: mdl-34201219

ABSTRACT

Background: The therapeutically important DNA repair gene O6-methylguanine DNA methyltransferase (MGMT) is silenced by promoter methylation in human brain cancers. The co-players/regulators associated with this process and the subsequent progression of MGMT gene transcription beyond the non-coding exon 1 are unknown. As a follow-up to our recent finding of a predicted second promoter mapped proximal to the exon 2 [Int. J. Mol. Sci.2021, 22(5), 2492], we addressed its significance in MGMT transcription. Methods: RT-PCR, RT q-PCR, and nuclear run-on transcription assays were performed to compare and contrast the transcription rates of exon 1 and exon 2 of the MGMT gene in glioblastoma cells. Results: Bioinformatic characterization of the predicted MGMT exon 2 promoter showed several consensus TATA box and INR motifs and the absence of CpG islands in contrast to the established TATA-less, CpG-rich, and GAF-bindable exon 1 promoter. RT-PCR showed very weak MGMT-E1 expression in MGMT-proficient SF188 and T98G GBM cells, compared to active transcription of MGMT-E2. In the MGMT-deficient SNB-19 cells, the expression of both exons remained weak. The RT q-PCR revealed that MGMT-E2 and MGMT-E5 expression was about 80- to 175-fold higher than that of E1 in SF188 and T98G cells. Nuclear run-on transcription assays using bromo-uridine immunocapture followed by RT q-PCR confirmed the exceptionally lower and higher transcription rates for MGMT-E1 and MGMT-E2, respectively. Conclusions: The results provide the first evidence for transcriptional pausing at the promoter 1- and non-coding exon 1 junction of the human MGMT gene and its activation/elongation through the protein-coding exons 2 through 5, possibly mediated by a second promoter. The findings offer novel insight into the regulation of MGMT transcription in glioma and other cancer types.


Subject(s)
Brain Neoplasms/metabolism , DNA Modification Methylases/genetics , DNA Repair Enzymes/genetics , Gene Expression Regulation, Neoplastic , Glioblastoma/metabolism , Tumor Suppressor Proteins/genetics , Brain Neoplasms/genetics , Cell Line, Tumor , DNA Methylation , DNA Modification Methylases/metabolism , DNA Repair Enzymes/metabolism , Exons , Glioblastoma/genetics , Humans , Promoter Regions, Genetic , Tumor Suppressor Proteins/metabolism
11.
Methods Mol Biol ; 2193: 85-96, 2021.
Article in English | MEDLINE | ID: mdl-32808261

ABSTRACT

Lymphangiogenesis, the formation of lymphatic vessels from preexisting ones, is an important process in wound-healing physiology. Deregulation of lymphangiogenesis and lymphatic vascular remodeling have been implicated in a range of inflammatory conditions, such as lymphedema, lymphadenopathy, tumor growth, and cancer metastasis. Any attempt in understanding various parameters of the lymphangiogenic process and developing desirable therapeutic targets requires recapitulating these conditions in in vivo models. One pitfall with some experimental models is the absence of immune response, an important regulatory factor for lymphangiogenesis. We overcome this issue by using immune competent mice. In this chapter, by using Angiopoietin-2 (Ang2), a protein that belongs to the Ang/Tie signaling pathway, we describe the ear sponge assay with important adaptations, highlighting a reproducible and quantitative tool for assessment of in vivo lymphangiogenesis.


Subject(s)
Biological Assay/methods , Ear/physiopathology , Lymphangiogenesis/physiology , Lymphatic Vessels/physiology , Angiopoietin-2/genetics , Animals , Ear/surgery , Humans , Immunity/immunology , Immunity/physiology , Lymphangiogenesis/genetics , Lymphangiogenesis/immunology , Lymphatic Vessels/immunology , Mice , Signal Transduction/genetics , Vascular Remodeling/genetics , Vascular Remodeling/immunology , Vascular Remodeling/physiology , Wound Healing/genetics , Wound Healing/physiology
12.
Bioconjug Chem ; 21(2): 203-7, 2010 Feb 17.
Article in English | MEDLINE | ID: mdl-20063878

ABSTRACT

Although avidin-mediated intracellular delivery of oligonucleotides or proteins has been shown before, the efficacy studies are lacking. Here, we tested the effectiveness of avidin for delivery of a cytochrome P450 reductase (CPR) antisense oligo in rat liver epithelial cells. A phosphorodiamidate morpholino oligo (PMO) against CPR was biotinylated using four reagents with short, cleavable, or long linkers, followed by conjugation with avidin. The dose-inhibitory response of the unmodified PMO in the presence of a transfection reagent (Endoporter, EP) and the effectiveness of the EP-assisted and avidin-assisted delivery of biotinylated PMOs were tested by Western blot analysis. Additionally, in a preliminary study, the avidin-biotin PMO with a long linker was also tested in vivo in rats. The biotinylated oligos were at least as effective as the unmodified oligo. Whereas the avidin conjugate of biotinylated PMO with the short linker was ineffective, those with the long linkers showed significant reductions in CPR protein expression. Finally, the in vivo study showed modest, but significant, reductions in CPR activity. In conclusion, these studies show for the first time that avidin-mediated intracellular delivery of biotinylated oligos can effectively knock down target genes in vitro, depending on the length of the linker. Additionally, the avidin-biotin approach may be of potential value for in vivo gene knockdown.


Subject(s)
Avidin/chemistry , Biotin/chemistry , Drug Carriers/chemistry , Drug Carriers/metabolism , Morpholines/chemistry , Morpholines/metabolism , NADPH-Ferrihemoprotein Reductase/genetics , Animals , Avidin/metabolism , Biotin/metabolism , Biotinylation , Cell Line , Liver/cytology , Liver/metabolism , Male , Morpholinos , RNA, Messenger/genetics , Rats , Rats, Sprague-Dawley
13.
Cells ; 9(7)2020 07 01.
Article in English | MEDLINE | ID: mdl-32630235

ABSTRACT

There is a desperate need for novel and efficacious chemotherapeutic strategies for human brain cancers. There are abundant molecular alterations along the p53 and MDM2 pathways in human glioma, which play critical roles in drug resistance. The present study was designed to evaluate the in vitro and in vivo antitumor activity of a novel brain-penetrating small molecule MDM2 degrader, termed SP-141. In a panel of nine human glioblastoma and medulloblastoma cell lines, SP-141, as a single agent, potently killed the brain tumor-derived cell lines with IC50 values ranging from 35.8 to 688.8 nM. Treatment with SP-141 resulted in diminished MDM2 and increased p53 and p21cip1 levels, G2/M cell cycle arrest, and marked apoptosis. In intracranial xenograft models of U87MG glioblastoma (wt p53) and DAOY medulloblastoma (mutant p53) expressing luciferase, treatment with SP-141 caused a significant 4- to 9-fold decrease in tumor growth in the absence of discernible toxicity. Further, combination treatment with a low dose of SP-141 (IC20) and temozolomide, a standard anti-glioma drug, led to synergistic cell killing (1.3- to 31-fold) in glioma cell lines, suggesting a novel means for overcoming temozolomide resistance. Considering that SP-141 can be taken up by the brain without the need for any special delivery, our results suggest that SP-141 should be further explored for the treatment of tumors of the central nervous system, regardless of the p53 status of the tumor.


Subject(s)
Antineoplastic Agents/pharmacology , Brain Neoplasms/metabolism , Glioblastoma/metabolism , Indoles/pharmacology , Medulloblastoma/metabolism , Molecular Targeted Therapy/methods , Proto-Oncogene Proteins c-mdm2/antagonists & inhibitors , Pyridines/pharmacology , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/therapeutic use , Antineoplastic Combined Chemotherapy Protocols/pharmacology , Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Apoptosis/drug effects , Brain Neoplasms/drug therapy , Cell Line, Tumor , Cell Movement , Cyclin-Dependent Kinase Inhibitor p21/metabolism , G2 Phase Cell Cycle Checkpoints/drug effects , Glioblastoma/drug therapy , Humans , Indoles/therapeutic use , Inhibitory Concentration 50 , Medulloblastoma/drug therapy , Mice , Mice, Inbred NOD , Mice, Nude , Proto-Oncogene Proteins c-mdm2/metabolism , Pyridines/therapeutic use , Temozolomide/pharmacology , Temozolomide/therapeutic use , Xenograft Model Antitumor Assays
14.
Proteomics ; 9(10): 2776-87, 2009 May.
Article in English | MEDLINE | ID: mdl-19405034

ABSTRACT

RNF2, a member of polycomb group (PcG) proteins, is involved in chromatin remodeling. However, mechanisms that regulate RNF2 function are unknown. To identify such mechanisms, RNF2 was expressed in HEK-293 cells and analyzed by 2-D electrophoresis. RNF2 was resolved into at least seven protein spots, migrating toward the lower pI from its expected pI of 6.38, suggesting that RNF2 undergoes post-translational modifications. Western blotting indicated that majority of these RNF2 spots contained phosphoserine(s), which were completely dephosphorylated upon treatment with a phosphatase. SB203580, a specific inhibitor of p38 MAPK, inhibited RNF2 phosphorylation at one site. On the other hand, PD98059, an inhibitor of MEK1/2, inhibited majority of the phosphorylation events in RNF2. Mass spectrometry analysis identified that RNF2 expressed in Sf9 insect cells undergoes co-translational excision of (1)Met coupled to N-acetylation of (2)Ser, and phosphorylation of (41)Ser. Interestingly, (41)Ser is a predicted p38/MAPK phosphorylation site, consistent with the loss of phosphorylation induced by SB203580. Further analysis indicated that RNF2 phosphorylation differentially modulates the expression of transcription factors and histone 2B acetylation. These results provide first evidence for phosphorylation of RNF2, and suggest that the mitogen activated protein kinases including p38 MAPK and ERK1/2 regulate growth, stress response, differentiation and other cellular processes, through phosphorylation of RNF2.


Subject(s)
DNA-Binding Proteins/metabolism , Extracellular Signal-Regulated MAP Kinases/metabolism , MAP Kinase Signaling System , Repressor Proteins/metabolism , Ubiquitin-Protein Ligases/metabolism , p38 Mitogen-Activated Protein Kinases/metabolism , Cell Line , DNA-Binding Proteins/genetics , Electrophoresis, Gel, Two-Dimensional , Gene Expression Regulation , Humans , Phosphorylation , Phosphoserine/metabolism , Polycomb Repressive Complex 1 , Polycomb-Group Proteins , Protein Processing, Post-Translational , Proteomics , Repressor Proteins/genetics , Tandem Mass Spectrometry , Ubiquitin-Protein Ligases/genetics
15.
Int J Oncol ; 34(3): 597-608, 2009 Mar.
Article in English | MEDLINE | ID: mdl-19212664

ABSTRACT

The ability of aspirin to trigger apoptosis in cancer cells is well known and is consistent with the clinical and epidemiological evidence on its chemopreventive effects in curtailing epithelial cancers, including breast cancer. We hypothesized that the anticancer effects of aspirin may involve acetylation of the tumor suppressor protein p53, a known regulator of apoptosis. In the present study, we determined if aspirin at the physiologically achievable concentration of 100 microM acetylates p53 and modulates the expression of p21CIP1, a protein involved in cell cycle arrest, and Bax, a pro-apoptotic protein. Using MDA-MB-231 human breast cancer cells, we demonstrate that aspirin at 100 microM concentration markedly acetylated the p53 protein, which was primarily localized to the nucleus. Aspirin induced p21CIP1 protein levels in a transient fashion in contrast to the sustained induction of Bax. The induction of p21CIP1 protein levels began at 3 h and was maximal at 6-8 h; however, it decreased to control levels by 30 h. In contrast, the anticancer drug, camptothecin (CPT) induced a steady accumulation of p21CIP1 protein. Remarkably, when cells were co-treated with aspirin and CPT, p21CIP1 levels were drastically downregulated, and this phenomenon was observed in many cancer cell lines. Incubation of recombinant p21 with cytoplasmic extracts from aspirin-treated cells caused its degradation suggesting the involvement of proteases in the disappearance of p21CIP1. Consistent with this data, aspirin decreased the survival of CPT-treated cells and greatly increased the extent of apoptosis. Our observation that aspirin has the ability to inhibit p21CIP1 after its initial induction has important implications in chemotherapy, and suggests its potential use to increase the efficacy of anticancer agents.


Subject(s)
Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Aspirin/pharmacology , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Camptothecin/pharmacology , Cyclin-Dependent Kinase Inhibitor p21/biosynthesis , Acetylation/drug effects , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Camptothecin/antagonists & inhibitors , Cell Nucleus/metabolism , Cyclin-Dependent Kinase Inhibitor p21/antagonists & inhibitors , Cyclin-Dependent Kinase Inhibitor p21/genetics , DNA, Neoplasm/metabolism , Drug Synergism , Humans , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Tumor Cells, Cultured , Tumor Suppressor Protein p53/biosynthesis , Tumor Suppressor Protein p53/metabolism , bcl-2-Associated X Protein/biosynthesis
16.
Int J Oncol ; 35(1): 193-204, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19513568

ABSTRACT

Previously, we reported that acetaminophen (APAP) showed selective toxicity towards melanoma cell lines. In the current study, we investigated further the role of tyrosinase in APAP toxicity in SK-MEL-28 melanoma cells in the presence of a short hairpin RNA (shRNA) plasmid, silencing tyrosinase gene. Results from tyrosinase shRNA experiments showed that APAP led to negligible toxicity in shRNA plasmid-treated cells. It was also found that APAP selectively caused escalation in reactive oxygen species (ROS) formation and intracellular GSH (ICG) depletion in melanocytic human SK-MEL-28 and murine B16-F0 melanoma cells that express functional tyrosinase whereas it lacked significant effects on ROS formation and ICG in amelanotic C32 melanoma cells that do not express functional tyrosinase. These findings suggest that tyrosinase plays a major role in APAP selective induced toxicity in melanocytic melanoma cell lines. Furthermore, the in vivo efficacy and toxicity of APAP in the skin melanoma tumor model in mice was investigated. Mice receiving APAP at 60, 80, 100 and 300 mg/kg/day, day 7 through 13 post melanoma cell inoculation demonstrated tumor size growth inhibition by 7+/-14, 30+/-17, 45+/-11 and 57+/-3%, respectively. Mice receiving APAP day 1 through 13 post melanoma cell inoculation showed tumor size growth inhibition by 11+/-7, 33+/-9, 36+/-20 and 44+/-28%, respectively.


Subject(s)
Acetaminophen/pharmacology , Antineoplastic Agents/pharmacology , Melanoma, Experimental/drug therapy , Skin Neoplasms/drug therapy , Acetaminophen/toxicity , Animals , Antineoplastic Agents/toxicity , Ascorbic Acid/metabolism , Cell Line, Tumor , Cell Survival/drug effects , Dose-Response Relationship, Drug , Glutathione/metabolism , Humans , Inhibitory Concentration 50 , Kidney/drug effects , Kidney/metabolism , Lipid Peroxidation/drug effects , Liver/drug effects , Liver/metabolism , Male , Melanoma, Experimental/enzymology , Melanoma, Experimental/genetics , Melanoma, Experimental/pathology , Mice , Mice, Inbred C57BL , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/metabolism , NAD/metabolism , Oxidation-Reduction , Phenacetin/metabolism , RNA Interference , Reactive Oxygen Species/metabolism , Skin Neoplasms/enzymology , Skin Neoplasms/genetics , Skin Neoplasms/pathology , Sulfhydryl Compounds/metabolism , Time Factors
17.
Int J Mol Med ; 24(1): 103-10, 2009 Jul.
Article in English | MEDLINE | ID: mdl-19513542

ABSTRACT

Angiotensin II is well implicated in neointimal proliferation and the resulting restenosis, however, the mechanisms involved remain unclear. The type 2 angiotensin II (AT2) receptor, largely unexpressed in the adult vasculature, however, appears at significant levels after vascular injury. To investigate the specific contribution of AT2 receptor and the interplay of the angiotensin system to neointima, we engineered rat vascular smooth muscle cells (VSMCs) to express the AT2 receptor in a tetracycline-regulated system. Several VSMC clones resistant to both hygromycin and G418 were selected, many of which showed high, but regulatable levels of AT2R expression within 48 h of doxycycline (Dox) exposure. In untransfected VSMCs and stable transfectants with no AT2R induction, Ang II significantly increased the expression of matrix metalloproteinase 2 (MMP-2), which is linked to neointimal growth. However, induction of AT2R by Dox addition markedly decreased MMP-2 levels (P<0.01) and this downregulation was further promoted by CV-11974, a specific antagonist of AT1 receptor. In contrast, the PD123319 compound, which selectively curtails the AT2 receptor, reversed the inhibition caused by CV-11974. We conclude that Ang II enhances the MMP-2 expression via AT1R, and that enforces AT2R inhibited the same. These data confirm that AT2R functions to downregulate the effects elicited by Ang II + AT1R signaling and point to the role of MMP and extracellular matrix in vascular injury. The findings provide fresh experimental approaches to prevent or control restenosis through transduction of VSMCs expressing optimal levels of AT2R.


Subject(s)
Angiotensin II/metabolism , Endothelium, Vascular/metabolism , Matrix Metalloproteinase 2/metabolism , Muscle, Smooth, Vascular/metabolism , Receptor, Angiotensin, Type 2/metabolism , Angiotensin II Type 1 Receptor Blockers/pharmacology , Angiotensin II Type 2 Receptor Blockers , Animals , Benzimidazoles/pharmacology , Biphenyl Compounds , Endothelium, Vascular/drug effects , Imidazoles/pharmacology , Matrix Metalloproteinase Inhibitors , Muscle, Smooth, Vascular/drug effects , Pyridines/pharmacology , Rats , Receptor, Angiotensin, Type 2/genetics , Signal Transduction , Tetrazoles/pharmacology
18.
Sci Rep ; 9(1): 8577, 2019 06 12.
Article in English | MEDLINE | ID: mdl-31189950

ABSTRACT

The Near-infrared Fluorescence (NIRF) molecular imaging of cancer is known to be superior in sensitivity, deeper penetration, and low phototoxicity compared to other imaging modalities. In view of an increased need for efficient and targeted imaging agents, we synthesized a NAD(P)H quinone oxidoreductase 1 (NQO1)-activatable NIR fluorescent probe (NIR-ASM) by conjugating dicyanoisophorone (ASM) fluorophore with the NQO1 substrate quinone propionic acid (QPA). The probe remained non-fluorescent until activation by NQO1, whose expression is largely limited to malignant tissues. With a large Stokes shift (186 nm) and a prominent near-infrared emission (646 nm) in response to NQO1, NIR-ASM was capable of monitoring NQO1 activity in vitro and in vivo with high specificity and selectivity. We successfully employed the NIR-ASM to differentiate cancer cells from normal cells based on NQO1 activity using fluorescence microscopy and flow cytometry. Chemical and genetic approaches involving the use of ES936, a specific inhibitor of NQO1 and siRNA and gene transfection procedures unambiguously demonstrated NQO1 to be the sole target activating the NIR-ASM in cell cultures. NIR-ASM was successfully used to detect and image the endogenous NQO1 in three live tumor-bearing mouse models (A549 lung cancer, Lewis lung carcinoma, and MDMAMB 231 xenografts) with a high signal-to-low noise ratiometric NIR fluorescence response. When the NQO1-proficient A549 tumors and NQO1-deficient MDA-MB-231 tumors were developed in the same animal, only the A549 malignancies activated the NIR-ASM probe with a strong signal. Because of its high sensitivity, rapid activation, tumor selectivity, and nontoxic properties, the NIR-ASM appears to be a promising agent with clinical applications.


Subject(s)
Fluorescent Dyes/chemistry , NAD(P)H Dehydrogenase (Quinone)/metabolism , Neoplasm Proteins/metabolism , Neoplasms, Experimental/diagnostic imaging , Neoplasms, Experimental/metabolism , A549 Cells , Animals , Female , Flow Cytometry , Heterografts , Humans , Mice , Mice, Nude , Microscopy, Fluorescence , NAD(P)H Dehydrogenase (Quinone)/genetics , Neoplasm Proteins/genetics , Neoplasm Transplantation , Neoplasms, Experimental/genetics
19.
Int J Oncol ; 32(1): 135-44, 2008 Jan.
Article in English | MEDLINE | ID: mdl-18097552

ABSTRACT

P-glycoprotein (Pgp), a plasma membrane (PM) glycoprotein, is responsible for the development of multidrug resistance. The mechanism by which Pgp is targeted to the PM is not defined. To identify proteins that influence Pgp trafficking, we utilized the yeast two-hybrid analysis procedure, which identified a new isoform of endoplasmic reticulum (ER)-bound Bap29, termed Bap29varP, as an interacting protein with the N-terminus of Pgp. The drug-resistant human breast cancer MCF-7 (MCF-7/Adr(R)) cells express both Bap29varP and approximately 170 kDa Pgp, which are however absent in the drug-sensitive MCF-7 cells. When Bap29varP was overexpressed in MCF-7/Adr(R) cells, Pgp was predominantly localized in the ER and intracellular vesicles, suggesting Bap29varP influences Pgp trafficking. When Pgp was expressed in MCF-7 cells, it was exclusively found in the ER with a molecular mass of approximately 160 kDa slightly smaller than that of the molecular mass of Pgp expressed in MCF-7/Adr(R) cells. On the other hand, when Pgp was expressed in Bap29varP-containing human colon adenocarcinoma HT-29 cells, it was localized at the PM. These findings together suggest that Bap29varP acts as an essential chaperone, influencing the processing and trafficking of Pgp to the cell surface.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/analysis , Membrane Proteins/physiology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Amino Acid Sequence , Base Sequence , Cell Line, Tumor , Cell Membrane/chemistry , Cell Membrane/metabolism , Glycosylation , Humans , Membrane Proteins/analysis , Membrane Proteins/chemistry , Membrane Proteins/genetics , Molecular Sequence Data , Protein Isoforms , Protein Processing, Post-Translational , Receptors, Estrogen/analysis
20.
Oncol Rep ; 19(3): 587-94, 2008 Mar.
Article in English | MEDLINE | ID: mdl-18288388

ABSTRACT

Galectins play a key role in oncogenic processes. Although several galectins are known, their relative expression at the mRNA and protein levels, the subcellular localization, and their relationship to the oncogenic manifestation remains unclear. Herein we report a comprehensive characterization of the expression of major galectins in human breast cancer (drug-sensitive MCF-7 and drug-resistant MCF-7/Adr(R)), colon cancer (HCT-116 and HT-29), and glioma (T98G) cell lines, as these cells are common model systems for studying oncogenic processes. The expected approximately 14.5 kDa galectin-1, predominantly cytosolic, was detected in the cancer and normal cell lines. Notably, two different molecular forms of galectin-1 with molecular masses of approximately 13.5 and 15 kDa were detected in T98G cells, the latter being in the extracellular medium, perhaps a result of post-translational processing. Immunocytochemistry indicated that the extracellular galectin-1 bound to the cell surface was punctated in appearance, suggesting that it was bound to specific receptors. Immunohistological studies indicated that metastasizing carcinomas express high levels of galectin-1. On the other hand, galectin-3 was readily detectable in all cancer cell lines but undetectable in normal cell lines, indicating that galectin-3 is a cancer-specific biomarker protein. Galectin-3 was a cytosolic protein but was not detected in the extracellular medium, indicating that cancer cells do not secrete this galectin. Finally, despite the RT-PCR analysis suggesting the presence of two transcripts of galectin-8 in all cancer cell lines, the corresponding approximately 36 kDa protein was only detectable in the nuclear and cytosolic fractions upon cell fractionation. Notably, a different molecular form of galectin-8 of approximately 18 kDa was immunoprecipitated from the extracellular media, suggesting that the secreted galectin-8 undergoes post-translational processing. These results highlight the expression of galectins in different molecular forms in cancers, warranting caution in interpreting the results of functional studies of individual galectins, particularly because these proteins function redundantly in cancer pathways.


Subject(s)
Galectins/analysis , Galectins/metabolism , Neoplasms/metabolism , Breast Neoplasms/chemistry , Breast Neoplasms/genetics , Breast Neoplasms/metabolism , Cell Line , Cell Line, Tumor , Colonic Neoplasms/chemistry , Colonic Neoplasms/genetics , Colonic Neoplasms/metabolism , Galectin 1/chemistry , Galectin 1/genetics , Galectin 1/metabolism , Galectin 3/chemistry , Galectin 3/genetics , Galectin 3/metabolism , Galectins/chemistry , Galectins/genetics , Humans , Immunohistochemistry , Neoplasms/chemistry , Neoplasms/genetics , Reverse Transcriptase Polymerase Chain Reaction
SELECTION OF CITATIONS
SEARCH DETAIL