Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Sci Adv ; 10(18): eadi3653, 2024 May 03.
Article in English | MEDLINE | ID: mdl-38691599

ABSTRACT

Manipulating the nanostructure of materials is critical for numerous applications in electronics, magnetics, and photonics. However, conventional methods such as lithography and laser writing require cleanroom facilities or leave residue. We describe an approach to creating atomically sharp line defects in hexagonal boron nitride (hBN) at room temperature by direct optical phonon excitation with a mid-infrared pulsed laser from free space. We term this phenomenon "unzipping" to describe the rapid formation and growth of a crack tens of nanometers wide from a point within the laser-driven region. Formation of these features is attributed to the large atomic displacement and high local bond strain produced by strongly driving the crystal at a natural resonance. This process occurs only via coherent phonon excitation and is highly sensitive to the relative orientation of the crystal axes and the laser polarization. Its cleanliness, directionality, and sharpness enable applications such as polariton cavities, phonon-wave coupling, and in situ flake cleaving.

2.
Nat Commun ; 14(1): 7685, 2023 Nov 24.
Article in English | MEDLINE | ID: mdl-38001087

ABSTRACT

Polar crystals can be driven into collective oscillations by optical fields tuned to precise resonance frequencies. As the amplitude of the excited phonon modes increases, novel processes scaling non-linearly with the applied fields begin to contribute to the dynamics of the atomic system. Here we show two such optical nonlinearities that are induced and enhanced by the strong phonon resonance in the van der Waals crystal hexagonal boron nitride (hBN). We predict and observe large sub-picosecond duration signals due to four-wave mixing (FWM) during resonant excitation. The resulting FWM signal allows for time-resolved observation of the crystal motion. In addition, we observe enhancements of third-harmonic generation with resonant pumping at the hBN transverse optical phonon. Phonon-induced nonlinear enhancements are also predicted to yield large increases in high-harmonic efficiencies beyond the third.

3.
NPJ Breast Cancer ; 7(1): 60, 2021 May 26.
Article in English | MEDLINE | ID: mdl-34040000

ABSTRACT

The effect of scheduling of targeted therapy combinations on drug resistance is underexplored in triple-negative breast cancer (TNBC). TNBC constitutes heterogeneous cancer cell populations the composition of which can change dynamically during treatment resulting in the selection of resistant clones with a fitness advantage. We evaluated crizotinib (ALK/MET inhibitor) and navitoclax (ABT-263; Bcl-2/Bcl-xL inhibitor) combinations in a large design consisting of 696 two-cycle sequential and concomitant treatment regimens with varying treatment dose, duration, and drug holiday length over a 26-day period in MDA-MB-231 TNBC cells and found that patterns of resistance depend on the schedule and sequence in which the drugs are given. Further, we tracked the clonal dynamics and mechanisms of resistance using DNA-integrated barcodes and single-cell RNA sequencing. Our study suggests that longer formats of treatment schedules in vitro screening assays are required to understand the effects of resistance and guide more realistically in vivo and clinical studies.

4.
Opt Lett ; 46(8): 1824-1827, 2021 Apr 15.
Article in English | MEDLINE | ID: mdl-33857079

ABSTRACT

We measure the third-order nonlinear optical response of various dielectrics and semiconductors using the spectrally resolved two-beam coupling method at 2.3 µm, 3.5 µm, 4.5 µm, and 8.3 µm. These materials include fused silica, sapphire, calcium fluoride, magnesium fluoride, zinc sulphide, and zinc selenide. We compare our results with previous literature results and theoretically expected values using two-band model theory. The dispersion of the nonlinear refractive index n2 over this wavelength range is found to be negligible.

5.
Cancers (Basel) ; 12(9)2020 Sep 08.
Article in English | MEDLINE | ID: mdl-32911681

ABSTRACT

Cancer cells employ various defense mechanisms against drug-induced cell death. Investigating multi-omics landscapes of cancer cells before and after treatment can reveal resistance mechanisms and inform new therapeutic strategies. We assessed the effects of navitoclax, a BCL2 family inhibitor, on the transcriptome, methylome, chromatin structure, and copy number variations of MDA-MB-231 triple-negative breast cancer (TNBC) cells. Cells were sampled before treatment, at 72 h of exposure, and after 10-day drug-free recovery from treatment. We observed transient alterations in the expression of stress response genes that were accompanied by corresponding changes in chromatin accessibility. Most of these changes returned to baseline after the recovery period. We also detected lasting alterations in methylation states and genome structure that suggest permanent changes in cell population composition. Using single-cell analyses, we identified 2350 genes significantly upregulated in navitoclax-resistant cells and derived an 18-gene navitoclax resistance signature. We assessed the navitoclax-response-predictive function of this signature in four additional TNBC cell lines in vitro and in silico in 619 cell lines treated with 251 different drugs. We observed a drug-specific predictive value in both experiments, suggesting that this signature could help guiding clinical biomarker studies involving navitoclax.

6.
Opt Lett ; 44(23): 5888-5891, 2019 Dec 01.
Article in English | MEDLINE | ID: mdl-31774805

ABSTRACT

Ionization occurs ubiquitously in intense laser-matter interaction and often leads to rapid decrease in laser intensity via plasma defocusing, shortening the effective interaction length of desired high-field processes. Refocusing of pulses may compensate for this adverse effect. However, it typically relies on Kerr-induced self-focusing and requires sufficiently high power. Here, we present simulations showing the refocusing of intense pulses with an initial Gaussian beam profile in atmospheric pressure gases at relatively low power. We attribute this refocusing to the formation of ring-structure plasmas. We find that tighter focusing leads to stronger refocusing, and the initial chirp of the pulse greatly affects its dynamics due to spatiotemporal coupling of focused broadband pulses. Our results highlight a novel aspect of complex pulse dynamics and can be relevant to applications involving tightly focused ultrafast Gaussian beams.

7.
Sci Rep ; 9(1): 14934, 2019 10 17.
Article in English | MEDLINE | ID: mdl-31624295

ABSTRACT

The goal of this study was to identify a novel target for antibody-drug conjugate (ADC) development in triple negative breast cancer (TNBC), which has limited treatment options, using gene expression datasets and in vitro siRNA/CRISPR and in vivo functional assays. We analyzed 4467 breast cancers and identified GABRP as top expressed gene in TNBC with low expression in most normal tissues. GABRP protein was localized to cell membrane with broad range of receptors/cell (815-53,714) and expressed by nearly half of breast cancers tissues. GABRP gene knockdown inhibited TNBC cell growth and colony formation in vitro and growth of MDA-MB-468 xenografts in nude mice. Commercially available anti-GABRP antibody (5-100 µg/ml) or de novo generated Fabs (20 µg/ml) inhibited TNBC cell growth in vitro. The same antibody conjugated to mertansine (DM1) also showed significant anticancer activity at nanomolar concentrations. Our results indicate that GABRP is a potential novel therapeutic target for ADC development.


Subject(s)
Antineoplastic Agents/pharmacology , GABA-A Receptor Antagonists/pharmacology , Immunoconjugates/pharmacology , Receptors, GABA-A/metabolism , Triple Negative Breast Neoplasms/drug therapy , Animals , Antineoplastic Agents/therapeutic use , Breast/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Proliferation/drug effects , Datasets as Topic , Drug Development , Female , GABA-A Receptor Antagonists/therapeutic use , Gene Expression Profiling , Gene Knockdown Techniques , Humans , Immunoconjugates/therapeutic use , Immunoglobulin Fab Fragments/pharmacology , Immunoglobulin Fab Fragments/therapeutic use , Maytansine/pharmacology , Maytansine/therapeutic use , Mice , Molecular Targeted Therapy/methods , Receptors, GABA-A/genetics , Triple Negative Breast Neoplasms/pathology , Xenograft Model Antitumor Assays
8.
Clin Epigenetics ; 10(1): 112, 2018 08 29.
Article in English | MEDLINE | ID: mdl-30157950

ABSTRACT

BACKGROUND: Age is one of the most important risk factors for developing breast cancer. However, age-related changes in normal breast tissue that potentially lead to breast cancer are incompletely understood. Quantifying tissue-level DNA methylation can contribute to understanding these processes. We hypothesized that occurrence of breast cancer should be associated with an acceleration of epigenetic aging in normal breast tissue. RESULTS: Ninety-six normal breast tissue samples were obtained from 88 subjects (breast cancer = 35 subjects/40 samples, unaffected = 53 subjects/53 samples). Normal tissue samples from breast cancer patients were obtained from distant non-tumor sites of primary mastectomy specimens, while samples from unaffected women were obtained from the Komen Tissue Bank (n = 25) and from non-cancer-related breast surgery specimens (n = 28). Patients were further stratified into four cohorts: age < 50 years with and without breast cancer and age ≥ 50 with and without breast cancer. The Illumina HumanMethylation450k BeadChip microarray was used to generate methylation profiles from extracted DNA samples. Data was analyzed using the "Epigenetic Clock," a published biomarker of aging based on a defined set of 353 CpGs in the human genome. The resulting age estimate, DNA methylation age, was related to chronological age and to breast cancer status. The DNAmAge of normal breast tissue was strongly correlated with chronological age (r = 0.712, p < 0.001). Compared to unaffected peers, breast cancer patients exhibited significant age acceleration in their normal breast tissue (p = 0.002). Multivariate analysis revealed that epigenetic age acceleration in the normal breast tissue of subjects with cancer remained significant after adjusting for clinical and demographic variables. Additionally, smoking was found to be positively correlated with epigenetic aging in normal breast tissue (p = 0.012). CONCLUSIONS: Women with luminal breast cancer exhibit significant epigenetic age acceleration in normal adjacent breast tissue, which is consistent with an analogous finding in malignant breast tissue. Smoking is also associated with epigenetic age acceleration in normal breast tissue. Further studies are needed to determine whether epigenetic age acceleration in normal breast tissue is predictive of incident breast cancer and whether this mediates the risk of chronological age on breast cancer risk.


Subject(s)
Breast Neoplasms/genetics , Breast/chemistry , CpG Islands , DNA Methylation , High-Throughput Nucleotide Sequencing/methods , Adult , Age Factors , Case-Control Studies , Epigenesis, Genetic , Female , Humans , Middle Aged , Sequence Analysis, DNA , Tissue Banks
9.
Opt Lett ; 43(13): 3112-3115, 2018 Jul 01.
Article in English | MEDLINE | ID: mdl-29957799

ABSTRACT

We demonstrate numerically and experimentally that intense pulses propagating in gas-filled capillaries can undergo localization in space and time due to strong plasma defocusing. This phenomenon can occur below or above the self-focusing threshold Pcr as a result of ionization-induced refraction that excites higher-order modes. The constructive interference of higher-order modes leads to spatiotemporal localization and resurgence of the intensity. Simulations show that this confinement is more prominent at shorter wavelength pulses and for smaller capillary diameters. Experiments with ultraviolet pulses show evidence that this ionization-induced refocusing appears below Pcr and thus represents a mechanism for spatiotemporal confinement without self-focusing.

10.
Oncotarget ; 8(13): 21733-21740, 2017 Mar 28.
Article in English | MEDLINE | ID: mdl-28423514

ABSTRACT

Luminal breast tumors have been classified into A and B subgroups, with the luminal A being associated with a more favorable clinical outcome. Unfortunately, luminal A tumors do not have a universally good prognosis. We used transcriptomic analyses using public datasets to evaluate the differential expression between normal breast tissue and breast cancer, focusing on upregulated genes included in cell cycle function. Association of selected genes with relapse free survival (RFS) and overall survival (OS) was performed using the KM Plotter Online Tool (http://www.kmplot.com). Seventy-seven genes were differentially expressed between normal and malignant breast tissue. Only five genes were associated with poor RFS and OS. The mitosis-related genes GTSE1, CDCA3, FAM83D and SMC4 were associated with poor outcome specifically in Luminal A tumors. The combination of FAM83D and CDCA3 for RFS and GTSE1 alone for OS showed the better prediction for clinical outcome. CDCA3 was amplified in 3.4% of the tumors, and FAM83D and SMC4 in 2.3% and 2.2%, respectively. In conclusion, we describe a set of genes that predict detrimental outcome in Luminal A tumors. These genes may have utility for stratification in trials of antimitotic agents or cytotoxic chemotherapy, or as candidates for direct target inhibition.


Subject(s)
Breast Neoplasms/genetics , Transcriptome , Breast Neoplasms/mortality , Breast Neoplasms/pathology , Disease-Free Survival , Female , Gene Expression Profiling , Humans , Kaplan-Meier Estimate
11.
Cancer Res ; 77(2): 566-578, 2017 01 15.
Article in English | MEDLINE | ID: mdl-27872098

ABSTRACT

Triple-negative breast cancer (TNBC) remains an aggressive disease without effective targeted therapies. In this study, we addressed this challenge by testing 128 FDA-approved or investigational drugs as either single agents or in 768 pairwise drug combinations in TNBC cell lines to identify synergistic combinations tractable to clinical translation. Medium-throughput results were scrutinized and extensively analyzed for sensitivity patterns, synergy, anticancer activity, and were validated in low-throughput experiments. Principal component analysis revealed that a fraction of all upregulated or downregulated genes of a particular targeted pathway could partly explain cell sensitivity toward agents targeting that pathway. Combination therapies deemed immediately tractable to translation included ABT-263/crizotinib, ABT-263/paclitaxel, paclitaxel/JQ1, ABT-263/XL-184, and paclitaxel/nutlin-3, all of which exhibited synergistic antiproliferative and apoptotic activity in multiple TNBC backgrounds. Mechanistic investigations of the ABT-263/crizotinib combination offering a potentially rapid path to clinic demonstrated RTK blockade, inhibition of mitogenic signaling, and proapoptotic signal induction in basal and mesenchymal stem-like TNBC. Our findings provide preclinical proof of concept for several combination treatments of TNBC, which offer near-term prospects for clinical translation. Cancer Res; 77(2); 566-78. ©2016 AACR.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/pharmacology , Drug Screening Assays, Antitumor/methods , Triple Negative Breast Neoplasms , Blotting, Western , Cell Line, Tumor , Cell Proliferation/drug effects , Drug Synergism , Female , Flow Cytometry , Humans , Immunoprecipitation , Principal Component Analysis
12.
J Bioenerg Biomembr ; 48(2): 153-68, 2016 Apr.
Article in English | MEDLINE | ID: mdl-25620271

ABSTRACT

The sphingolipid family of lipids modulate several cellular processes, including proliferation, cell cycle regulation, inflammatory signaling pathways, and cell death. Several members of the sphingolipid pathway have opposing functions and thus imbalances in sphingolipid metabolism result in deregulated cellular processes, which cause or contribute to diseases and disorders in humans. A key cellular process regulated by sphingolipids is apoptosis, or programmed cell death. Sphingolipids play an important role in both extrinsic and intrinsic apoptotic pathways depending on the stimuli, cell type and cellular response to the stress. During mitochondrial-mediated apoptosis, multiple pathways converge on mitochondria and induce mitochondrial outer membrane permeabilization (MOMP). MOMP results in the release of intermembrane space proteins such as cytochrome c and Apaf1 into the cytosol where they activate the caspases and DNases that execute cell death. The precise molecular components of the pore(s) responsible for MOMP are unknown, but sphingolipids are thought to play a role. Here, we review evidence for a role of sphingolipids in the induction of mitochondrial-mediated apoptosis with a focus on potential underlying molecular mechanisms by which altered sphingolipid metabolism indirectly or directly induce MOMP. Data available on these mechanisms is reviewed, and the focus and limitations of previous and current studies are discussed to present important unanswered questions and potential future directions.


Subject(s)
Apoptosis/physiology , Mitochondria/metabolism , Mitochondrial Membranes/metabolism , Sphingolipids/metabolism , Animals , Apoptotic Protease-Activating Factor 1/genetics , Apoptotic Protease-Activating Factor 1/metabolism , Humans , Mitochondria/genetics , Permeability , Sphingolipids/genetics
13.
Biochim Biophys Acta ; 1848(10 Pt A): 2374-84, 2015 Oct.
Article in English | MEDLINE | ID: mdl-26215742

ABSTRACT

Ceramide is a bioactive sphingolipid involved in mitochondrial-mediated apoptosis. Our data suggest that ceramides directly regulate a key initiation step in apoptosis: mitochondrial outer membrane permeabilization (MOMP). MOMP allows release of intermembrane space proteins to the cytosol, inducing the execution of the cell. Ceramides form channels in planar phospholipid membranes and outer membranes of isolated mitochondria, channels large enough to facilitate passage of proteins released during MOMP. Bcl-xL inhibits MOMP in vivo and inhibits the formation of ceramide channels in vitro. However the significance of Bcl-xL's regulation of ceramide channel formation within cells was untested. We engineered Bcl-xL point mutations that specifically affect the interaction between ceramide and Bcl-xL to probe the mechanism of ceramide channel regulation and the role of ceramide channels in apoptosis. Using these mutants and fluorescently-labeled ceramide, we identified the hydrophobic groove on Bcl-xL as the critical ceramide binding site and regulator of ceramide channel formation. Bcl-xL mutants with weakened interaction with ceramide also have reduced ability to interfere with ceramide channel formation. Some mutants have similar altered ability to inhibit both ceramide and Bax channel formation, whereas others act differentially, suggesting distinct but overlapping binding sites. To probe the relative importance of these channels in apoptosis, Bcl-xL mutant proteins were stably expressed in Bcl-xL deficient cells. Weakening the inhibition of either Bax or ceramide channels decreased the ability of Bcl-xL to protect cells from apoptosis in a stimulus-dependent manner. These studies provide the first in vivo evidence for the role of ceramide channels in MOMP.


Subject(s)
Ceramides/chemistry , Ceramides/metabolism , Mitochondria, Liver/physiology , Mitochondrial Membranes/physiology , bcl-X Protein/chemistry , bcl-X Protein/metabolism , Animals , Apoptosis/physiology , Binding Sites , Cell Membrane Permeability/physiology , Cells, Cultured , Humans , Mice , Mitochondria, Liver/ultrastructure , Mitochondrial Membranes/ultrastructure , Molecular Dynamics Simulation , Protein Binding , Rats , Rats, Sprague-Dawley
14.
Biochim Biophys Acta ; 1841(11): 1571-80, 2014 Nov.
Article in English | MEDLINE | ID: mdl-25195822

ABSTRACT

Mutants of tumor suppressor p53 not only lose the activity in genome stabilizing and in tumor suppression, but also exhibit oncogenic function in cancer cells. Most efforts in restoring p53 biological activity focus on either altering mutant-protein conformation or introducing an exogenous p53 gene into cells to eliminate p53-mutant cancer cells. Being different from these, we report that ceramide can restore the expression of wild-type p53 and induce p53-dependent apoptosis in deletion-mutant cancer cells. We show that endogenous long-carbon chain ceramide species (C16- to C24-ceramides) and exogenous C6-ceramide, rather than other sphingolipids, restore wild-type mRNA (intact exon-5), phosphorylated protein (Ser15 in exon-5) of p53, and p53-responsive proteins, including p21 and Bax, in ovarian cancer cells, which predominantly express a deleted exon-5 of p53 mutant before treatments. Consequently, the restored p53 sensitizes these p53-mutant cancer cells to DNA damage-induced growth arrest and apoptosis. Furthermore, we elucidate that ceramide activates protein phosphatase-1, and then the dephosphorylated serine/arginine-rich splicing-factor 1 (SRSF1) is translocated to the nucleus, thus promoting pre-mRNA splicing preferentially to wild-type p53 expression. These findings disclose an unrecognized mechanism that pre-mRNA splicing dysfunction can result in p53 deletion-mutants. Ceramide through SRSF1 restores wild-type p53 expression versus deletion-mutant and leads cancer cells to apoptosis. This suggests that heterozygous deletion-mutants of p53 can be restored in posttranscriptional level by using epigenetic approaches.

15.
Int J Oncol ; 39(2): 425-31, 2011 Aug.
Article in English | MEDLINE | ID: mdl-21617856

ABSTRACT

Drug resistance causes treatment failure in approximately 50% of breast cancer patients with chemotherapy. Overexpression of glucosylceramide synthase (GCS) confers drug resistance in cancer cells, and suppression of GCS sensitizes cancers to chemotherapy in preclinical studies. Thus, GCS becomes a potential target to reverse drug resistance; however, little is known about GCS expression levels in normal tissues and whether GCS overexpression is associated with metastatic cancers. Herewith, we report our studies in GCS expression levels and breast cancer from patients. GCS levels were analyzed using cancer profiling arrays, breast cancer histo-arrays and quantitative RT-PCR in tumor tissues. We found that breast (18 exp. index) and other hormone-dependent organs (testis, cervix, ovary, prostate) displayed the lowest levels of GCS mRNA, whereas liver (52 exp. index) and other organs (kidney, bladder, stomach) displayed the highest levels of GCS. GCS mRNA levels were significantly elevated in tumors of breast, cervix, rectum and small intestine, as compared to each paired normal tissue. In mammary tissue, GCS overexpression was detected in breast cancers with metastasis, but not in benign fibroadenoma or primary tumors. GCS overexpression was coincident with HER2 expression (γ2=0.84) in ER-negative breast adenocarcinoma. In tumor specimens, GCS mRNA was elevated by 4-fold and significantly associated with stage III (5/7), lymph node-positive (7/8) and estrogen receptor-positive breast cancers (7/9). GCS expression was significantly and selectively elevated in breast cancer, in particular in metastatic disease. GCS overexpression was highly associated with ER-positive and HER2-positive breast cancer with metastasis. Although a small study, these data suggest that GCS may be a prognostic indicator and potential target for the treatment of chemotherapy-refractory breast cancer.


Subject(s)
Breast Neoplasms/enzymology , Drug Resistance, Neoplasm/physiology , Gene Expression Regulation, Neoplastic , Glucosyltransferases/genetics , Glucosyltransferases/metabolism , Adult , Aged , Breast Neoplasms/genetics , Cell Line, Tumor , Disease Progression , Drug Resistance, Neoplasm/genetics , Female , Gene Expression Profiling , HL-60 Cells , HeLa Cells , Humans , K562 Cells , Middle Aged , Neoplasm Staging , RNA, Messenger/genetics , Receptor, ErbB-2/genetics , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Tissue Array Analysis
16.
Cancer Res ; 71(6): 2276-85, 2011 Mar 15.
Article in English | MEDLINE | ID: mdl-21278235

ABSTRACT

Tumor suppressor p53 plays an essential role in protecting cells from malignant transformation by inducing cell-cycle arrest and apoptosis. Mutant p53 that is detected in more than 50% of cases of cancers loses its role in suppression of tumors but gains in oncogenic function. Strategies to convert mutant p53 into wild-type p53 have been suggested for cancer prevention and treatment, but they face a variety of challenges. Here, we report an alternative approach that involves suppression of glucosylceramide synthase (GCS), an enzyme that glycosylates ceramide and blunts its proapoptotic activity in cancer cells. Human ovarian cancer cells expressing mutant p53 displayed resistance to apoptosis induced by DNA damage. We found that GCS silencing sensitized these mutant p53 cells to doxorubicin but did not affect the sensitivity of cells with wild-type p53. GCS silencing increased the levels of phosphorylated p53 and p53-responsive genes, including p21(Waf1/Cip1), Bax, and Puma, consistent with a redirection of the mutant p53 cells to apoptosis. Reactivated p53-dependent apoptosis was similarly verified in p53-mutant tumors where GCS was silenced. Inhibition of ceramide synthase with fumonisin B1 prevented p53 reactivation induced by GCS silencing, whereas addition of exogenous C6-ceramide reactivated p53 function in p53-mutant cells. Our findings indicate that restoring active ceramide to cells can resuscitate wild-type p53 function in p53-mutant cells, offering preclinical support for a novel type of mechanism-based therapy in the many human cancers harboring p53 mutations.


Subject(s)
Apoptosis , Glucosyltransferases/metabolism , Neoplasms/metabolism , Tumor Suppressor Protein p53/metabolism , Animals , Antineoplastic Agents/pharmacology , Apoptosis Regulatory Proteins/genetics , Apoptosis Regulatory Proteins/metabolism , Blotting, Western , Cell Cycle , Cell Line, Tumor , Ceramides/metabolism , Cyclin-Dependent Kinase Inhibitor p21/genetics , Cyclin-Dependent Kinase Inhibitor p21/metabolism , Dose-Response Relationship, Drug , Drug Resistance, Neoplasm/drug effects , Drug Resistance, Neoplasm/genetics , Glucosyltransferases/antagonists & inhibitors , Glucosyltransferases/genetics , Glycosphingolipids/metabolism , Glycosylation/drug effects , Humans , Mice , Mice, Nude , Mutation , Neoplasms/pathology , Neoplasms/therapy , Oligonucleotides/genetics , Oligonucleotides/pharmacology , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Tumor Suppressor Protein p53/genetics , Xenograft Model Antitumor Assays/methods
17.
Prog Lipid Res ; 50(1): 104-14, 2011 Jan.
Article in English | MEDLINE | ID: mdl-20970453

ABSTRACT

Sphingolipids including glycosphingolipids have myriad effects on cell functions and affect cancer in aspects of tumorigenesis, metastasis and tumor response to treatments. Bioactive ones like ceramide, sphingosine 1-phosphate and globotriaosylceramide initiate and process cellular signaling to alter cell behaviors immediately responding to oncogenic stress or treatment challenges. Recent studies pinpoint that sphingolipid-mediated gene expression has long and profound impacts on cancer cells, and these play crucial roles in tumor progression and in treatment outcome. More than 10 sphingolipids and glycosphingolipids selectively mediate expressions of approximately 50 genes including c-myc, p21, c-fos, telomerase reverse transcriptase, caspase-9, Bcl-x, cyclooxygenase-2, matrix metalloproteinases, integrins, Oct-4, glucosylceramide synthase and multidrug-resistant gene 1. By diverse functions of these genes, sphingolipids enduringly affect cellular processes of mitosis, apoptosis, migration, stemness of cancer stem cells and cellular resistance to therapies. Mechanistic studies indicate that sphingolipids regulate particular gene expression by modulating phosphorylation and acetylation of proteins that serve as transcription factors (ß-catenin, Sp1), repressor of transcription (histone H3), and regulators (SRp30a) in RNA splicing. Disclosing molecular mechanisms by which sphingolipids selectively regulate particular gene expression, instead of other relevant ones, requires understanding of the exact roles of individual lipid instead of a group, the signaling pathways that are implicated in and interaction with proteins or other lipids in details. These studies not only expand our knowledge of sphingolipids, but can also suggest novel targets for cancer treatments.


Subject(s)
Gene Expression Regulation, Neoplastic , Neoplasms/genetics , Neoplasms/metabolism , Sphingolipids/metabolism , Animals , Humans , Neoplasms/drug therapy , Neoplasms/pathology
18.
Int J Pharm ; 400(1-2): 251-9, 2010 Nov 15.
Article in English | MEDLINE | ID: mdl-20816930

ABSTRACT

In this study, solid lipid nanoparticles (SLN) loaded with MBO-asGCS oligonucleotide were prepared, characterized and evaluated for cytotoxicity against NCI/ADR-RES human ovary cancer cells. Two types of cetyltrimethyl ammonium bromide (CTAB) stabilized SLN, with or without ceramide VI, were prepared by mixed homogenization/ultrasonication technique. Complexes were characterized for size, zeta-potential, and stability in biorelevant media and against DNaseI activity. Binding and release studies were further confirmed by gel electrophoresis. Cytotoxicity of the SLN against NCI/ADR-RES cells was evaluated by quantizing ATP. SLN with ceramide VI had lower particle size (74.6 nm) with improved stability in RPMI media when compared to reference SLN without ceramide VI (167.16 nm). Both SLN however had similar cytotoxicity profile with an optimum binding at CTAB to MBO-asGCS ratio of 6:1. Blank SLN, and free MBO-asGCS in the presence and absence of free doxorubicin had insignificant effect on the viability of NCI/ADR-RES cells. However, when cells were concurrently treated with MBO-asGCS loaded SLN and free doxorubicin, cell viability significantly decreased to approximately 12%. These results suggested that SLN enhanced internalization and uptake of MBO-asGCS oligonucleotide, which led to the downregulation of GCS and subsequently reversing the resistance of the cells to doxorubicin.


Subject(s)
Drug Carriers/chemistry , Drug Resistance, Neoplasm/drug effects , Glucosyltransferases/genetics , Lipids/chemistry , Nanoparticles/chemistry , Oligonucleotides, Antisense/chemistry , Antibiotics, Antineoplastic/chemistry , Antibiotics, Antineoplastic/pharmacology , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival/drug effects , Chemistry, Pharmaceutical , Doxorubicin/chemistry , Doxorubicin/pharmacology , Drug Resistance, Multiple/drug effects , Female , Humans , Ovarian Neoplasms , Particle Size
19.
Mol Cancer ; 9: 145, 2010 Jun 11.
Article in English | MEDLINE | ID: mdl-20540746

ABSTRACT

BACKGROUND: Drug resistance is the outcome of multiple-gene interactions in cancer cells under stress of anticancer agents. MDR1 overexpression is most commonly detected in drug-resistant cancers and accompanied with other gene alterations including enhanced glucosylceramide synthase (GCS). MDR1 encodes for P-glycoprotein that extrudes anticancer drugs. Polymorphisms of MDR1 disrupt the effects of P-glycoprotein antagonists and limit the success of drug resistance reversal in clinical trials. GCS converts ceramide to glucosylceramide, reducing the impact of ceramide-induced apoptosis and increasing glycosphingolipid (GSL) synthesis. Understanding the molecular mechanisms underlying MDR1 overexpression and how it interacts with GCS may find effective approaches to reverse drug resistance. RESULTS: MDR1 and GCS were coincidently overexpressed in drug-resistant breast, ovary, cervical and colon cancer cells; silencing GCS using a novel mixed-backbone oligonucleotide (MBO-asGCS) sensitized these four drug-resistant cell lines to doxorubicin. This sensitization was correlated with the decreased MDR1 expression and the increased doxorubicin accumulation. Doxorubicin treatment induced GCS and MDR1 expression in tumors, but MBO-asGCS treatment eliminated "in-vivo" growth of drug-resistant tumor (NCI/ADR-RES). MBO-asGCS suppressed the expression of MDR1 with GCS and sensitized NCI/ADR-RES tumor to doxorubicin. The expression of P-glycoprotein and the function of its drug efflux of tumors were decreased by 4 and 8 times after MBO-asGCS treatment, even though this treatment did not have a significant effect on P-glycoprotein in normal small intestine. GCS transient transfection induced MDR1 overexpression and increased P-glycoprotein efflux in dose-dependent fashion in OVCAR-8 cancer cells. GSL profiling, silencing of globotriaosylceramide synthase and assessment of signaling pathway indicated that GCS transfection significantly increased globo series GSLs (globotriaosylceramide Gb3, globotetraosylceramide Gb4) on GSL-enriched microdomain (GEM), activated cSrc kinase, decreased beta-catenin phosphorylation, and increased nuclear beta-catenin. These consequently increased MDR1 promoter activation and its expression. Conversely, MBO-asGCS treatments decreased globo series GSLs (Gb3, Gb4), cSrc kinase and nuclear beta-catenin, and suppressed MDR-1 expression in dose-dependent pattern. CONCLUSION: This study demonstrates, for the first time, that GCS upregulates MDR1 expression modulating drug resistance of cancer. GSLs, in particular globo series GSLs mediate gene expression of MDR1 through cSrc and beta-catenin signaling pathway.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/genetics , Drug Resistance, Neoplasm , Gene Expression Regulation/physiology , Glucosyltransferases/physiology , Proto-Oncogene Proteins pp60(c-src)/metabolism , Signal Transduction/physiology , beta Catenin/metabolism , ATP Binding Cassette Transporter, Subfamily B , Antineoplastic Agents/pharmacology , Doxorubicin/pharmacology , Gene Silencing , Glucosyltransferases/genetics , Humans
20.
Biochem Pharmacol ; 80(1): 72-9, 2010 Jul 01.
Article in English | MEDLINE | ID: mdl-20298675

ABSTRACT

Multidrug resistance is a major impediment to the success of cancer chemotherapy. The overproduced P-glycoprotein that extrudes anticancer drugs from cells, is the most common mechanism detected in multidrug-resistant cancers. Direct measurement of cellular efflux of tumors in vivo, rather than estimation of MDR1 mRNA and P-glycoprotein levels in samples stored or embedded, can functionally characterize the mechanism of drug resistance and determine the choice of anticancer drugs for cancer patients. Herewith, we introduce a new approach to directly determine P-glycoprotein efflux of tumors. Employing Flutax-2 (Oregon green-488 paclitaxel) and fluorescence spectrophotometry, this method has successfully measured cellular transportability including efflux and accumulation in diverse cancer cell lines, tumors and other tissues with high reproducibility. With this method, we have quantitatively determined cellular efflux that is correlated with P-glycoprotein levels and the reversal effects of agents in cell lines of breast, ovarian, cervical and colon cancers, and in tumor-bearing mice. It has sensitively detected these alterations of P-glycoprotein efflux in approximately 5mg tumor or other tissues with high confidence. This direct and quick functional assessment has a potential to determine drug resistance in different types of cancers after surgical resection. Further validation of this method in clinic settings for the diagnosis of drug resistance purpose is needed.


Subject(s)
ATP Binding Cassette Transporter, Subfamily B, Member 1/physiology , Antineoplastic Agents/pharmacology , Drug Resistance, Multiple/drug effects , Neoplasms/drug therapy , Taxoids/pharmacology , ATP Binding Cassette Transporter, Subfamily B, Member 1/metabolism , Cell Line, Tumor , Colonic Neoplasms/therapy , Dose-Response Relationship, Drug , Humans , Kinetics , RNA, Messenger/analysis , RNA, Messenger/metabolism
SELECTION OF CITATIONS
SEARCH DETAIL
...