Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 21
Filter
Add more filters










Publication year range
1.
Nat Cancer ; 5(3): 433-447, 2024 Mar.
Article in English | MEDLINE | ID: mdl-38286827

ABSTRACT

Liver metastasis (LM) confers poor survival and therapy resistance across cancer types, but the mechanisms of liver-metastatic organotropism remain unknown. Here, through in vivo CRISPR-Cas9 screens, we found that Pip4k2c loss conferred LM but had no impact on lung metastasis or primary tumor growth. Pip4k2c-deficient cells were hypersensitized to insulin-mediated PI3K/AKT signaling and exploited the insulin-rich liver milieu for organ-specific metastasis. We observed concordant changes in PIP4K2C expression and distinct metabolic changes in 3,511 patient melanomas, including primary tumors, LMs and lung metastases. We found that systemic PI3K inhibition exacerbated LM burden in mice injected with Pip4k2c-deficient cancer cells through host-mediated increase in hepatic insulin levels; however, this circuit could be broken by concurrent administration of an SGLT2 inhibitor or feeding of a ketogenic diet. Thus, this work demonstrates a rare example of metastatic organotropism through co-optation of physiological metabolic cues and proposes therapeutic avenues to counteract these mechanisms.


Subject(s)
Liver Neoplasms , Proto-Oncogene Proteins c-akt , Humans , Mice , Animals , Proto-Oncogene Proteins c-akt/metabolism , Phosphatidylinositol 3-Kinases , Signal Transduction , Insulin , Phosphotransferases (Alcohol Group Acceptor)/metabolism
2.
Cancer Med ; 12(18): 19033-19046, 2023 09.
Article in English | MEDLINE | ID: mdl-37596773

ABSTRACT

BACKGROUND: Patient-reported data can improve quality of healthcare delivery and patient outcomes. Moffitt Cancer Center ("Moffitt") administers the Electronic Patient Questionnaire (EPQ) to collect data on demographics, including sexual orientation and gender identity (SOGI), medical history, cancer risk factors, and quality of life. Here we investigated differences in EPQ completion by demographic and cancer characteristics. METHODS: An analysis including 146,142 new adult patients at Moffitt in 2009-2020 was conducted using scheduling, EPQ and cancer registry data. EPQ completion was described by calendar year and demographics. Logistic regression was used to estimate associations between demographic/cancer characteristics and EPQ completion. More recently collected information on SOGI were described. RESULTS: Patient portal usage (81%) and EPQ completion rates (79%) were consistently high since 2014. Among patients in the cancer registry, females were more likely to complete the EPQ than males (odds ratio [OR] = 1.17, 95% confidence interval [CI] = 1.14-1.20). Patients ages 18-64 years were more likely to complete the EPQ than patients aged ≥65. Lower EPQ completion rates were observed among Black or African American patients (OR = 0.59, 95% CI = 0.56-0.63) as compared to Whites and among patients whose preferred language was Spanish (OR = 0.40, 95% CI = 0.36-0.44) or another language as compared to English. Furthermore, patients with localized (OR = 1.16, 95% CI = 1.12-1.19) or regional (OR = 1.16, 95% CI = 1.12-1.20) cancer were more likely to complete the EPQ compared to those with metastatic disease. Less than 3% of patients self-identified as being lesbian, gay, or bisexual and <0.1% self-identified as transgender, genderqueer, or other. CONCLUSIONS: EPQ completion rates differed across demographics highlighting opportunities for targeted process improvement. Healthcare organizations should evaluate data acquisition methods to identify potential disparities in data completeness that can impact quality of clinical care and generalizability of self-reported data.


Subject(s)
Gender Identity , Neoplasms , Adult , Humans , Male , Female , Quality of Life , Sexual Behavior , Neoplasms/epidemiology , Neoplasms/therapy , Patient Reported Outcome Measures
3.
Cancer Discov ; 13(4): 1002-1025, 2023 04 03.
Article in English | MEDLINE | ID: mdl-36715544

ABSTRACT

KRAS is the most frequently mutated oncogene in human lung adenocarcinomas (hLUAD), and activating mutations frequently co-occur with loss-of-function mutations in TP53 or STK11/LKB1. However, mutation of all three genes is rarely observed in hLUAD, even though engineered comutation is highly aggressive in mouse lung adenocarcinoma (mLUAD). Here, we provide a mechanistic explanation for this difference by uncovering an evolutionary divergence in the regulation of triosephosphate isomerase (TPI1). In hLUAD, TPI1 activity is regulated via phosphorylation at Ser21 by the salt inducible kinases (SIK) in an LKB1-dependent manner, modulating flux between the completion of glycolysis and production of glycerol lipids. In mice, Ser21 of TPI1 is a Cys residue that can be oxidized to alter TPI1 activity without a need for SIKs or LKB1. Our findings suggest this metabolic flexibility is critical in rapidly growing cells with KRAS and TP53 mutations, explaining why the loss of LKB1 creates a liability in these tumors. SIGNIFICANCE: Utilizing phosphoproteomics and metabolomics in genetically engineered human cell lines and genetically engineered mouse models (GEMM), we uncover an evolutionary divergence in metabolic regulation within a clinically relevant genotype of human LUAD with therapeutic implications. Our data provide a cautionary example of the limits of GEMMs as tools to study human diseases such as cancers. This article is highlighted in the In This Issue feature, p. 799.


Subject(s)
Adenocarcinoma of Lung , Lung Neoplasms , Triose-Phosphate Isomerase , Animals , Humans , Mice , Adenocarcinoma of Lung/genetics , Adenocarcinoma of Lung/metabolism , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Mutation , Protein Serine-Threonine Kinases/genetics , Protein Serine-Threonine Kinases/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Triose-Phosphate Isomerase/genetics , Triose-Phosphate Isomerase/metabolism
4.
Cancer Discov ; 11(10): 2456-2473, 2021 10.
Article in English | MEDLINE | ID: mdl-33947663

ABSTRACT

APOBEC3 enzymes are cytosine deaminases implicated in cancer. Precisely when APOBEC3 expression is induced during cancer development remains to be defined. Here we show that specific APOBEC3 genes are upregulated in breast ductal carcinoma in situ, and in preinvasive lung cancer lesions coincident with cellular proliferation. We observe evidence of APOBEC3-mediated subclonal mutagenesis propagated from TRACERx preinvasive to invasive non-small cell lung cancer (NSCLC) lesions. We find that APOBEC3B exacerbates DNA replication stress and chromosomal instability through incomplete replication of genomic DNA, manifested by accumulation of mitotic ultrafine bridges and 53BP1 nuclear bodies in the G1 phase of the cell cycle. Analysis of TRACERx NSCLC clinical samples and mouse lung cancer models revealed APOBEC3B expression driving replication stress and chromosome missegregation. We propose that APOBEC3 is functionally implicated in the onset of chromosomal instability and somatic mutational heterogeneity in preinvasive disease, providing fuel for selection early in cancer evolution. SIGNIFICANCE: This study reveals the dynamics and drivers of APOBEC3 gene expression in preinvasive disease and the exacerbation of cellular diversity by APOBEC3B through DNA replication stress to promote chromosomal instability early in cancer evolution.This article is highlighted in the In This Issue feature, p. 2355.


Subject(s)
APOBEC Deaminases/genetics , Breast Neoplasms/genetics , Carcinoma, Ductal/genetics , Carcinoma, Non-Small-Cell Lung/genetics , Lung Neoplasms/genetics , Animals , Cell Line, Tumor , Chromosomal Instability , DNA Replication , Female , Humans , Mice
5.
Anal Methods ; 13(19): 2238-2247, 2021 05 20.
Article in English | MEDLINE | ID: mdl-33929476

ABSTRACT

Effective staining of peripheral blood smears by increasing contrast of intracellular components and biomarkers is essential for the accurate characterization, diagnosis, and monitoring of various diseases such as malaria. To assess the potential for automation of stained whole human blood smears at the point-of-care (POC), brightfield and fluorescence staining protocols were adapted for smears generated in channels of pumpless microchannels and compared to a standard glass smear. A 3× concentration Giemsa brightfield staining solutions (10, 33, and 50% dilution), and Acridine Orange fluorescence staining solutions (12 µg mL-1) were evaluated with human blood smears containing malaria parasites within a microfluidic channel. Giemsa staining at 33% dilution showed an optimal combination of contrast and preservation of cellular morphology, while 50% dilutions showed significant cellular crenation and 10% dilutions did not show desired contrast in brightfield imaging. Fluorescence staining at 12 µg mL-1 using Acridine Orange showed clear separability between the fluorescent intensities of the malaria parasites and that of the red blood cells (RBCs) and background. However, compared to glass smears, these exhibited reduced signal intensity as well as inverted contrast of RBCs and background. These results demonstrate that peripheral thin blood smears generated in pumpless microfluidic can be successfully stained in-channel with a simple, one-step procedure to permit brightfield and fluorescence imaging.


Subject(s)
Malaria , Microfluidics , Acridine Orange , Erythrocytes , Humans , Malaria/diagnosis , Staining and Labeling
6.
Cancer Metab ; 9(1): 12, 2021 Mar 24.
Article in English | MEDLINE | ID: mdl-33762003

ABSTRACT

BACKGROUND: Fructose is an abundant source of carbon and energy for cells to use for metabolism, but only certain cell types use fructose to proliferate. Tumor cells that acquire the ability to metabolize fructose have a fitness advantage over their neighboring cells, but the proteins that mediate fructose metabolism in this context are unknown. Here, we investigated the determinants of fructose-mediated cell proliferation. METHODS: Live cell imaging and crystal violet assays were used to characterize the ability of several cell lines (RKO, H508, HepG2, Huh7, HEK293T (293T), A172, U118-MG, U87, MCF-7, MDA-MB-468, PC3, DLD1 HCT116, and 22RV1) to proliferate in fructose (i.e., the fructolytic ability). Fructose metabolism gene expression was determined by RT-qPCR and western blot for each cell line. A positive selection approach was used to "train" non-fructolytic PC3 cells to utilize fructose for proliferation. RNA-seq was performed on parental and trained PC3 cells to find key transcripts associated with fructolytic ability. A CRISPR-cas9 plasmid containing KHK-specific sgRNA was transfected in 293T cells to generate KHK-/- cells. Lentiviral transduction was used to overexpress empty vector, KHK, or GLUT5 in cells. Metabolic profiling was done with seahorse metabolic flux analysis as well as LC/MS metabolomics. Cell Titer Glo was used to determine cell sensitivity to 2-deoxyglucose in media containing either fructose or glucose. RESULTS: We found that neither the tissue of origin nor expression level of any single gene related to fructose catabolism determine the fructolytic ability. However, cells cultured chronically in fructose can develop fructolytic ability. SLC2A5, encoding the fructose transporter, GLUT5, was specifically upregulated in these cells. Overexpression of GLUT5 in non-fructolytic cells enabled growth in fructose-containing media across cells of different origins. GLUT5 permitted fructose to flux through glycolysis using hexokinase (HK) and not ketohexokinase (KHK). CONCLUSIONS: We show that GLUT5 is a robust and generalizable driver of fructose-dependent cell proliferation. This indicates that fructose uptake is the limiting factor for fructose-mediated cell proliferation. We further demonstrate that cellular proliferation with fructose is independent of KHK.

7.
Soft Matter ; 17(15): 4133-4142, 2021 Apr 21.
Article in English | MEDLINE | ID: mdl-33735370

ABSTRACT

The ability to utilize extrusion-based, direct ink write (DIW) 3D printing to create silica-reinforced silicones with complex structures could expand their utility in industrial and biomedical applications. Sylgard 184, a common Pt-cure silicone, lacks the thixotropic behavior necessary for effective printing and its hydrophobicity renders cured structures susceptible to biofouling. Herein, we evaluated the efficacy of various PEO-silane amphiphiles (PEO-SAs) as thixotropic and surface modifying additives in Sylgard 184. Eight amphiphilic PEO-SAs of varying architecture (e.g. linear, star, and graft), crosslinkability, and PEO content were evaluated. Modified formulations were also prepared with additional amounts of silica filler, both hexamethyldisilazane (HMDS)-treated and dimethyldichlorosilane (DiMeDi)-treated types. Numerous PEO-SA modified silicone formulations demonstrated effective water-driven surface hydrophilicity that was generally diminished with the addition of HMDS-treated silica filler. While increased yield stress was observed for PEO-SA modified silicones with added HMDS-treated filler, none achieved the initial target for 3D printing (>1000 Pa). Only the formulations containing the DiMeDi-treated filler (17.3 wt%) were able to surpass this value. These formulations were then tested for their thixotropic properties and all surpassed the targets for recovered storage modulus (G') (>1000 Pa) and loss factor (<0.8). In particular, the triblock linear PEO-SA produced exceptionally high recovered G', low loss factor, and substantial water-driven restructuring to form a hydrophilic surface. Combined, these results demonstrate the potential of silicones modified with PEO-SA surface-modifying additives (SMAs) for extrusion-based, DIW 3D printing applications.

8.
Acta Biomater ; 121: 339-348, 2021 02.
Article in English | MEDLINE | ID: mdl-33271355

ABSTRACT

Silicones (i.e. crosslinked poly(dimethylsiloxane), PDMS) are commonly used material for microfluidic device fabrication. Nonetheless, due to the uncontrollable absorption of small hydrophobic molecules (<1 kDa) into the bulk, its applicability to cell-based drug assays and sensing applications has been limited. Here, we demonstrate the use of substrates made of silicones bulk modified with a poly(ethylene oxide) silane amphiphile (PEO-SA) to reduce hydrophobic small molecule sequestration for cell-based assays. Modified silicone substrates were generated with concentrations of 2 wt.%, 9 wt.% and, 14 wt.% PEO-SA. Incorporation of PEO-SA into the silicone bulk was assessed by FTIR analysis in addition to water contact angle analysis to evaluate surface hydrophobicity. Cell toxicity, absorption of small hydrophobic drugs, and cell response to hydrophobic molecules were also evaluated. Results showed that the incorporation of the PEO-SA into the silicone led to a reduction in water contact angle from 114° to as low as 16° that was stable for at least three months. The modified silicones showed viability values above 85% for NIH-3T3, MCF7, MDA-MB-468, and MDA-MB-231 cell lines. A drug response assay using tamoxifen and the MCF7 cell line showed full recovery of cell toxicity response when exposed to PDMS modified with 9 wt.% or 14 wt.% PEO-SA compared to tissue culture plastic. Therefore, our study supports the use of PEO-SA at concentrations of 9 wt.% or higher for enhanced surface wettability and reduced absorption of small hydrophobic molecules in PDMS-based platforms.


Subject(s)
Dimethylpolysiloxanes , Silicones , Hydrophobic and Hydrophilic Interactions , Polyethylene Glycols , Water , Wettability
9.
Macromol Biosci ; 20(12): e2000193, 2020 12.
Article in English | MEDLINE | ID: mdl-32812374

ABSTRACT

Surface-induced thrombosis is problematic in blood-contacting devices composed of silicones or polyurethanes (PUs). Poly(ethylene oxide)-silane amphiphiles (PEO-SA) are previously shown effective as surface modifying additives (SMAs) in silicones for enhanced thromboresistance. This study investigates PEO-SAs as SMAs in a PU at various concentrations: 5, 10, 25, 50, and 100 µmol g-1 PU. PEO-SA modified PUs are evaluated for their mechanical properties, water-driven surface restructuring, and adhesion resistance against a human fibrinogen (HF) solution as well as whole human blood. Stability is assessed by monitoring hydrophilicity, water uptake, and mass loss following air- or aqueous-conditioning. PEO-SA modified PUs do not demonstrate plasticization, as evidenced by minimal changes in glass transition temperature, modulus, tensile strength, and percent strain at break. These also show a concentration-dependent increase in hydrophilicity that is sustained following air- and aqueous-conditioning for concentrations ≥25 µmol g-1 . Additionally, water uptake and mass loss are minimal at all concentrations. Although protein resistance is not enhanced versus an HF solution, PEO-SA modified PUs have significantly reduced protein adsorption and platelet adhesion from human blood at concentrations ≥10 µmol g-1 . Overall, this study demonstrates the versatility of PEO-SAs as SMAs in PU, which leads to enhanced and sustained hydrophilicity as well as thromboresistance.


Subject(s)
Biocompatible Materials/pharmacology , Platelet Adhesiveness/drug effects , Polyethylene Glycols/chemistry , Thrombosis/prevention & control , Adsorption/drug effects , Biocompatible Materials/chemistry , Fibrinogen/chemistry , Humans , Polyethylene Glycols/pharmacology , Polyurethanes/chemistry , Silanes/chemistry , Silanes/pharmacology , Silicones/chemistry , Surface Properties/drug effects , Tensile Strength , Thrombosis/pathology , Water/chemistry
10.
Cancer Discov ; 10(9): 1352-1373, 2020 09.
Article in English | MEDLINE | ID: mdl-32571778

ABSTRACT

A hallmark of metastasis is the adaptation of tumor cells to new environments. Metabolic constraints imposed by the serine and glycine-limited brain environment restrict metastatic tumor growth. How brain metastases overcome these growth-prohibitive conditions is poorly understood. Here, we demonstrate that 3-phosphoglycerate dehydrogenase (PHGDH), which catalyzes the rate-limiting step of glucose-derived serine synthesis, is a major determinant of brain metastasis in multiple human cancer types and preclinical models. Enhanced serine synthesis proved important for nucleotide production and cell proliferation in highly aggressive brain metastatic cells. In vivo, genetic suppression and pharmacologic inhibition of PHGDH attenuated brain metastasis, but not extracranial tumor growth, and improved overall survival in mice. These results reveal that extracellular amino acid availability determines serine synthesis pathway dependence, and suggest that PHGDH inhibitors may be useful in the treatment of brain metastasis. SIGNIFICANCE: Using proteomics, metabolomics, and multiple brain metastasis models, we demonstrate that the nutrient-limited environment of the brain potentiates brain metastasis susceptibility to serine synthesis inhibition. These findings underscore the importance of studying cancer metabolism in physiologically relevant contexts, and provide a rationale for using PHGDH inhibitors to treat brain metastasis.This article is highlighted in the In This Issue feature, p. 1241.


Subject(s)
Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Brain/pathology , Phosphoglycerate Dehydrogenase/antagonists & inhibitors , Animals , Antineoplastic Agents/pharmacology , Brain/metabolism , Brain Neoplasms/secondary , Cell Line, Tumor , Datasets as Topic , Drug Resistance, Neoplasm , Female , Gene Knockdown Techniques , Glycine/analysis , Glycine/metabolism , Humans , Metabolomics , Mice , Phosphoglycerate Dehydrogenase/genetics , Phosphoglycerate Dehydrogenase/metabolism , Proteomics , RNA-Seq , Serine/analysis , Serine/metabolism , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays
11.
ACS Biomater Sci Eng ; 6(4): 2029-2037, 2020 04 13.
Article in English | MEDLINE | ID: mdl-33455354

ABSTRACT

The antifouling properties of poly(ethylene oxide) (PEO)-silane amphiphiles as surface-modifying additives (SMAs) in a condensation cure silicone have been previously demonstrated against simple protein solutions. Comprising an oligo(dimethylsiloxane) tether (m = 13 or 30) and PEO segment (n = 8), sustained protein resistance was achieved even in the absence of a cross-linkable triethoxysilane group, particularly when comprising the longer tether. To probe their potential for thromboresistance, PEO-silane amphiphile SMAs were used to bulk-modify silicones and evaluated for adhesion resistance against whole human blood under both static and dynamic conditions. Both a cross-linkable (XL diblock, m = 13) and a non-cross-linkable (Diblock, m = 30) SMA were evaluated at various concentrations (5-50 µmol SMA/g silicone) in a condensation cure silicone. Under static conditions, silicones modified with either SMA at concentrations of 10 µmol/g or greater were effective in reducing adhesion of human fibrinogen and platelets. Dynamic testing further showed that modified silicones were able to reduce protein adsorption and thrombus formation. This occurred at 5 and 10 µmol/g for silicones modified with XL diblock, m = 13 and Diblock, m = 30 SMAs, respectively. Combined, these results indicate the effectiveness of PEO-silane amphiphiles as SMAs in silicone for improved thromboresistance.


Subject(s)
Silanes , Silicones , Adsorption , Humans , Polyethylene Glycols , Surface Properties
12.
Nat Rev Cancer ; 19(5): 271-282, 2019 05.
Article in English | MEDLINE | ID: mdl-30967651

ABSTRACT

Over the past century, the notion that vitamin C can be used to treat cancer has generated much controversy. However, new knowledge regarding the pharmacokinetic properties of vitamin C and recent high-profile preclinical studies have revived interest in the utilization of high-dose vitamin C for cancer treatment. Studies have shown that pharmacological vitamin C targets many of the mechanisms that cancer cells utilize for their survival and growth. In this Opinion article, we discuss how vitamin C can target three vulnerabilities many cancer cells share: redox imbalance, epigenetic reprogramming and oxygen-sensing regulation. Although the mechanisms and predictive biomarkers that we discuss need to be validated in well-controlled clinical trials, these new discoveries regarding the anticancer properties of vitamin C are promising to help identify patient populations that may benefit the most from high-dose vitamin C therapy, developing effective combination strategies and improving the overall design of future vitamin C clinical trials for various types of cancer.


Subject(s)
Ascorbic Acid/pharmacology , Ascorbic Acid/therapeutic use , Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Epigenesis, Genetic/drug effects , Humans , Oxidation-Reduction/drug effects
13.
Nature ; 553(7689): 467-472, 2018 01 25.
Article in English | MEDLINE | ID: mdl-29342134

ABSTRACT

Chromosomal instability is a hallmark of cancer that results from ongoing errors in chromosome segregation during mitosis. Although chromosomal instability is a major driver of tumour evolution, its role in metastasis has not been established. Here we show that chromosomal instability promotes metastasis by sustaining a tumour cell-autonomous response to cytosolic DNA. Errors in chromosome segregation create a preponderance of micronuclei whose rupture spills genomic DNA into the cytosol. This leads to the activation of the cGAS-STING (cyclic GMP-AMP synthase-stimulator of interferon genes) cytosolic DNA-sensing pathway and downstream noncanonical NF-κB signalling. Genetic suppression of chromosomal instability markedly delays metastasis even in highly aneuploid tumour models, whereas continuous chromosome segregation errors promote cellular invasion and metastasis in a STING-dependent manner. By subverting lethal epithelial responses to cytosolic DNA, chromosomally unstable tumour cells co-opt chronic activation of innate immune pathways to spread to distant organs.


Subject(s)
Chromosomal Instability , Cytosol/metabolism , DNA, Neoplasm/metabolism , Neoplasm Metastasis/genetics , Animals , Brain Neoplasms/genetics , Brain Neoplasms/pathology , Brain Neoplasms/secondary , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Breast Neoplasms/secondary , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line , Chromosomal Instability/genetics , Chromosome Segregation , Cytosol/enzymology , Female , Head and Neck Neoplasms/genetics , Head and Neck Neoplasms/pathology , Humans , Inflammation/genetics , Inflammation/metabolism , Membrane Proteins/metabolism , Mesoderm/metabolism , Mice , Micronuclei, Chromosome-Defective , NF-kappa B/metabolism , Nucleotidyltransferases/metabolism , Xenograft Model Antitumor Assays
14.
Polym Chem ; 8(34): 5239-5251, 2017 Sep 14.
Article in English | MEDLINE | ID: mdl-29104619

ABSTRACT

Silicones with improved water-driven surface hydrophilicity and anti-biofouling behavior were achieved when bulk-modified with poly(ethylene oxide) (PEO) -silane amphiphiles of varying siloxane tether length: α-(EtO)3Si-(CH2)2-oligodimethylsiloxane m -block-poly(ethylene oxide)8-OCH3 (m = 0, 4, 13, 17, 24, and 30). A PEO8-silane [α-(EtO)3Si-(CH2)3-PEO8-OCH3] served as a conventional PEO-silane control. To examine anti-biofouling behavior in the absence versus presence of water-driven surface restructuring, the amphiphiles and control were surface-grafted onto silicon wafers and used to bulk-modify a medical-grade silicone, respectively. While the surface-grafted PEO-control exhibited superior protein resistance, it failed to appreciably restructure to the surface-water interface of bulk-modified silicone and thus led to poor protein resistance. In contrast, the PEO-silane amphiphiles, while less protein-resistant when surface-grafted onto silicon wafers, rapidly and substantially restructured in bulk-modified silicone, exhibiting superior hydrophilicity and protein resistance. A reduction of biofilm for several strains of bacteria and a fungus was observed for silicones modified with PEO-silane amphiphiles. Longer siloxane tethers maintained surface restructuring and protein resistance while displaying the added benefit of increased transparency.

15.
ACS Macro Lett ; 6(9): 992-1000, 2017 Sep 19.
Article in English | MEDLINE | ID: mdl-35650885

ABSTRACT

Toward improving implantable medical devices as well as diagnostic performance, the development of polymeric biomaterials having resistance to proteins remains a priority. Herein, we highlight key strategies reported in the recent literature that have relied upon improvement of surface hydrophilicity via direct surface modification methods or with bulk modification using surface modifying additives (SMAs). These approaches have utilized a variety of techniques to incorporate the surface hydrophilization agent, including physisorption, hydrogel network formation, surface grafting, layer-by-layer (LbL) assembly and blending base polymers with SMAs. While poly(ethylene glycol) (PEG) remains the gold standard, new alternatives have emerged such as polyglycidols, poly(2-oxazoline)s (POx), polyzwitterions, and amphiphilic block copolymers. While these new strategies provide encouraging results, the need for improved correlation between in vitro and in vivo protein resistance is critical. This may be achieved by employing complex protein solutions as well as strides to enhance the sensitivity of protein adsorption measurements.

16.
Green Mater ; 5(1): 4-13, 2017 Mar.
Article in English | MEDLINE | ID: mdl-31673356

ABSTRACT

Surface modifying additives (SMAs), which may be readily blended into silicones to improve anti-fouling behavior, must have excellent surface migration potential and must not leach into the aqueous environment. In this work, we evaluated the efficacy of a series of poly(ethylene oxide) (PEO)-based SMA amphiphiles which varied in terms of crosslinkability, siloxane tether length (m) and diblock versus triblock architectures. Specifically, crosslinkable, diblock PEO-silane amphiphiles with two oligodimethylsiloxane (ODMS) tether lengths [(EtO)3Si-(CH2)3-ODMS m -PEO8, m = 13 and 30] were compared to analogous non-crosslinkable, diblock (H-Si-ODMS m -PEO8) and triblock (PEO8-ODMS m -PEO8) SMAs. Prior to water conditioning, while all modified silicone coatings exhibited a high degree of water-driven surface restructuring, that prepared with the non-crosslinkable diblock SMA (m = 13) was the most hydrophilic. After conditioning, all modified silicone coatings were similarly hydrophilic and remained highly protein resistant, with the exception of PEO8-ODMS 30 -PEO8. Notably, despite twice the PEO content, triblock SMAs were not superior to diblock SMAs. For diblock SMAs, it was shown that water uptake and leaching were also similar whether or not the SMA was crosslinkable.

17.
Curr Drug Targets ; 15(1): 2-16, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24387338

ABSTRACT

Cancer is a consequence of mutations in genes that control cell proliferation, differentiation and cellular homeostasis. These genes are classified into two categories: oncogenes and tumor suppressor genes. Together, overexpression of oncogenes and loss of tumor suppressors are the dominant driving forces for tumorigenesis. Hence, targeting oncogenes and tumor suppressors hold tremendous therapeutic potential for cancer treatment. In the last decade, the predominant cancer drug discovery strategy has relied on a traditional reductionist approach of dissecting molecular signaling pathways and designing inhibitors for the selected oncogenic targets. Remarkable therapies have been developed using this approach; however, targeting oncogenes is only part of the picture. Our understanding of the importance of tumor suppressors in preventing tumorigenesis has also advanced significantly and provides a new therapeutic window of opportunity. Given that tumor suppressors are frequently mutated, deleted, or silenced with loss-of-function, restoring their normal functions to treat cancer holds tremendous therapeutic potential. With the rapid expansion in our knowledge of cancer over the last several decades, developing effective anticancer regimens against tumor suppressor pathways has never been more promising. In this article, we will review the concept of tumor suppression, and outline the major therapeutic strategies and challenges of targeting tumor suppressor networks for cancer therapeutics.


Subject(s)
Genes, Tumor Suppressor , Neoplasms/therapy , Genetic Therapy , Humans , Neoplasms/genetics
18.
J Biol Chem ; 288(48): 34403-13, 2013 Nov 29.
Article in English | MEDLINE | ID: mdl-24129578

ABSTRACT

Hec1 and Nuf2, core components of the NDC80 complex, are essential for kinetochore-microtubule attachment and chromosome segregation. It has been shown that both Hec1 and Nuf2 utilize their coiled-coil domains to form a functional dimer; however, details of the consequential significance and structural requirements to form the dimerization interface have yet to be elucidated. Here, we showed that Hec1 required three contiguous heptad repeats from Leu-324 to Leu-352, but not the entire first coiled-coil domain, to ensure overall stability of the NDC80 complex through direct interaction with Nuf2. Substituting the hydrophobic core residues, Leu-331, Val-338, and Ile-345, of Hec1 with alanine completely eliminated Nuf2 binding and blocked mitotic progression. Moreover, unlike most coiled-coil proteins, where the buried positions are composed of hydrophobic residues, Hec1 possessed an unusual distribution of glutamic acid residues, Glu-334, Glu-341, and Glu-348, buried within the interior dimerization interface, which complement with three Nuf2 lysine residues: Lys-227, Lys-234, and Lys-241. Substituting these corresponding residues with alanine diminished the binding affinity between Hec1 and Nuf2, compromised NDC80 complex formation, and adversely affected mitotic progression. Taken together, these findings demonstrated that three buried glutamic acid-lysine pairs, in concert with hydrophobic interactions of core residues, provide the major specificity and stability requirements for Hec1-Nuf2 dimerization and NDC80 complex formation.


Subject(s)
Cell Cycle Proteins/genetics , Chromosome Segregation/genetics , Nuclear Proteins/genetics , Protein Multimerization/genetics , Cell Cycle Proteins/chemistry , Cytoskeletal Proteins , Gene Expression Regulation, Developmental , HeLa Cells , Humans , Hydrophobic and Hydrophilic Interactions , Kinetochores/chemistry , Microtubules/chemistry , Microtubules/genetics , Mitosis , Multiprotein Complexes , Nuclear Proteins/chemistry , Protein Structure, Tertiary/genetics
19.
Mol Biol Cell ; 22(19): 3584-94, 2011 Oct.
Article in English | MEDLINE | ID: mdl-21832156

ABSTRACT

The spindle assemble checkpoint (SAC) is critical for accurate chromosome segregation. Hec1 contributes to chromosome segregation in part by mediating SAC signaling and chromosome alignment. However, the molecular mechanism by which Hec1 modulates checkpoint signaling and alignment remains poorly understood. We found that Hec1 serine 165 (S165) is preferentially phosphorylated at kinetochores. Phosphorylated Hec1 serine 165 (pS165) specifically localized to kinetochores of misaligned chromosomes, showing a spatiotemporal distribution characteristic of SAC molecules. Expressing an RNA interference (RNAi)-resistant S165A mutant in Hec1-depleted cells permitted normal progression to metaphase, but accelerated the metaphase-to-anaphase transition. The S165A cells were defective in Mad1 and Mad2 localization to kinetochores, regardless of attachment status. These cells often entered anaphase with lagging chromosomes and elicited increased segregation errors and cell death. In contrast, expressing S165E mutant in Hec1-depleted cells triggered defective chromosome alignment and severe mitotic arrest associated with increased Mad1/Mad2 signals at prometaphase kinetochores. A small portion of S165E cells eventually bypassed the SAC but showed severe segregation errors. Nek2 is the primary kinase responsible for kinetochore pS165, while PP1 phosphatase may dephosphorylate pS165 during SAC silencing. Taken together, these results suggest that modifications of Hec1 S165 serve as an important mechanism in modulating SAC signaling and chromosome alignment.


Subject(s)
Chromosome Segregation , M Phase Cell Cycle Checkpoints , Nuclear Proteins/metabolism , Protein Serine-Threonine Kinases/metabolism , Amino Acid Sequence , Anaphase , Calcium-Binding Proteins/genetics , Calcium-Binding Proteins/metabolism , Cell Cycle Proteins/genetics , Cell Cycle Proteins/metabolism , Cell Line, Tumor , Cytoskeletal Proteins , Humans , Kinetochores/metabolism , Mad2 Proteins , Metaphase , Mitosis , Molecular Sequence Data , NIMA-Related Kinases , Nuclear Proteins/genetics , Phosphorylation , Protein Serine-Threonine Kinases/genetics , RNA Interference , Repressor Proteins/genetics , Repressor Proteins/metabolism , Signal Transduction/genetics
20.
J Biol Chem ; 286(34): 30097-106, 2011 Aug 26.
Article in English | MEDLINE | ID: mdl-21705324

ABSTRACT

Proper assembly of mitotic spindles requires Hice1, a spindle-associated protein. Hice1 possesses direct microtubule binding activity at its N-terminal region and contributes to intraspindle microtubule nucleation as a subunit of the Augmin complex. However, whether microtubule binding activity of Hice1 is modulated by mitotic regulators remains unexplored. Here, we found that Aurora-A kinase, a major mitotic kinase, specifically binds to and phosphorylates Hice1. We identified four serine/threonine clusters on Hice1 that can be phosphorylated by Aurora-A in vitro. Of the four clusters, the Ser/Thr-17-21 cluster was the most critical for bipolar spindle assembly, whereas other phospho-deficient point mutants had a minimal effect on spindle assembly. Immunostaining with a phospho-Ser-19/20 phospho-specific antibody revealed that phosphorylated Hice1 primarily localizes to spindle poles during prophase to metaphase but gradually diminishes after anaphase. Consistently, the phospho-mimic 17-21E mutant reduced microtubule binding activity in vitro and diminished localization to spindles in vivo. Furthermore, expression of the 17-21E mutant led to decreased association of Fam29a, an Augmin component, with spindles. On the other hand, expression of the phospho-deficient 17-21A mutant permitted intraspindle nucleation but delayed the separation of early mitotic spindle poles and the timely mitotic progression. Taken together, these results suggest that Aurora-A modulates the microtubule binding activity of Hice1 in a spatiotemporal manner for proper bipolar spindle assembly.


Subject(s)
Metaphase/physiology , Microtubule-Associated Proteins/metabolism , Microtubules/metabolism , Prophase/physiology , Protein Serine-Threonine Kinases/metabolism , Spindle Apparatus/metabolism , Aurora Kinases , Cell Line, Tumor , Humans , Microtubule-Associated Proteins/genetics , Microtubules/genetics , Mutation , Phosphorylation/physiology , Protein Serine-Threonine Kinases/genetics , Protein Structure, Tertiary , Spindle Apparatus/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...