Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 29
Filter
1.
Biomedicines ; 12(6)2024 Jun 20.
Article in English | MEDLINE | ID: mdl-38927582

ABSTRACT

BACKGROUND: Mesenchymal stromal/stem cells (MSCs) play a critical role in wound healing. Corlicyte® is an MSC product derived from allogeneic umbilical cord tissue donated under an institutional review board-approved protocol and processed in accordance with section 501(a)(2)(B) of the Federal Food, Drug, and Cosmetic Act. This open-label phase 1 trial was performed under a United States Food and Drug Administration Investigational New Drug Application to establish the safety and tolerability of Corlicyte® in patients with diabetes and chronic diabetic foot ulcer (DFU). METHODS: Escalating doses were applied topically twice a week for up to 8 weeks after ulcer debridement, wound photography, and measurement. Subjects were followed for 4 weeks after the treatment phase. Adverse events were assessed at every visit. RESULTS: Nine subjects in 2 dosing cohorts completed the trial. No subjects experienced a serious adverse reaction to Corlicyte® or the development of anti-human leukocyte antigen (HLA) antibodies. Sixty percentage of subjects in the lower dose cohort experienced ulcer closure by Day 70 of follow-up, while the mean ulcer size was reduced by 54-67% in the other subjects. CONCLUSIONS: Topical administration of Corlicyte®, a novel biologic therapy consisting of allogeneic umbilical cord lining MSCs, appeared safe and tolerable and resulted in a significant decrease in ulcer area, demonstrating its potential as a therapy for healing of chronic DFU.

2.
Qual Manag Health Care ; 31(1): 28-33, 2022.
Article in English | MEDLINE | ID: mdl-34724456

ABSTRACT

BACKGROUND AND OBJECTIVES: During its monthly morbidity and mortality conference (MMC), the University of Colorado Division of Cardiology reviewed a "near-miss" patient safety event involving the delayed completion of a Stat-priority (ie, statim, meaning high priority) electrocardiogram (ECG). Because critical and interprofessional stakeholders participated in the conference, we hypothesized that the MMC would be associated with reduced ECG completion times. METHODS: Data were collected for in-hospital ECGs performed at the University of Colorado Hospital between January 1, 2017, and June 30, 2018. An interrupted time series analysis was used to estimate the immediate and ongoing impact of the MMC (held on February 28, 2018) on ECG completion times, stratified by order priority (Stat, Now, or Routine). The percentage of delayed Stat-priority ECGs was analyzed as a secondary outcome. RESULTS: Before the MMC, ECG completion times were stable for all order priorities ( P > .2), but the proportion of delayed Stat-priority ECGs increased from 5% in January 2017 to 20% in February 2018 ( P < .01). The MMC was associated with an immediate reduction in average daily ECG completion times for Routine (-18.4 minutes, P = .03) and Now (-8 minutes, P = .024) priority ECGs. No reduction was seen for Stat ECGs ( P = .97), though the percentage of delayed Stat ECGs stopped increasing ( P = .63). In the post-MMC period, completion times for Routine-priority ECGs increased and approached pre-MMC levels. CONCLUSIONS: The MMC was associated with an immediate, but temporary, improvement in ECG completion times. Although the observed clinical benefit of the MMC is novel, these data support the need for more durable reforms to sustain initial improvements.

3.
Case Rep Cardiol ; 2021: 8365283, 2021.
Article in English | MEDLINE | ID: mdl-34447595

ABSTRACT

Acute coronary syndrome is a rare complication of vasculitis. We present a case of fulminant medium-vessel vasculitis, most likely PAN, complicated by STEMI and stroke, that was successfully treated with percutaneous revascularization, high-quality stroke care, and immunosuppression. This case highlights the importance of prompt diagnosis and treatment of vasculitis and the recognition of coronary and cerebral ischemia as potentially serious complications.

4.
Mol Cancer Res ; 16(2): 187-196, 2018 02.
Article in English | MEDLINE | ID: mdl-29117940

ABSTRACT

The ETS family transcription factor ETV4 is aberrantly expressed in a variety of human tumors and plays an important role in carcinogenesis through upregulation of relevant target gene expression. Here, it is demonstrated that ETV4 is overexpressed in pancreatic cancer tissues as compared with the normal pancreas, and is associated with enhanced growth and rapid cell-cycle progression of pancreatic cancer cells. ETV4 expression was silenced through stable expression of a specific short hairpin RNA (shRNA) in two pancreatic cancer cell lines (ASPC1 and Colo357), while it was ectopically expressed in BXPC3 cells. Silencing of ETV4 in ASPC1 and Colo357 cells reduced the growth by 55.3% and 38.9%, respectively, while forced expression of ETV4 in BXPC3 cells increased the growth by 46.8% in comparison with respective control cells. Furthermore, ETV4-induced cell growth was facilitated by rapid transition of cells from G1- to S-phase of the cell cycle. Mechanistic studies revealed that ETV4 directly regulates the expression of Cyclin D1 CCND1, a protein crucial for cell-cycle progression from G1- to S-phase. These effects on the growth and cell cycle were reversed by the forced expression of Cyclin D1 in ETV4-silenced pancreatic cancer cells. Altogether, these data provide the first experimental evidence for a functional role of ETV4 in pancreatic cancer growth and cell-cycle progression.Implications: The functional and mechanistic data presented here regarding ETV4 in pancreatic cancer growth and cell-cycle progression suggest that ETV4 could serve as a potential biomarker and novel target for pancreatic cancer therapy. Mol Cancer Res; 16(2); 187-96. ©2017 AACR.


Subject(s)
Adenovirus E1A Proteins/genetics , Adenovirus E1A Proteins/metabolism , Cyclin D1/genetics , Pancreatic Neoplasms/genetics , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Cell Cycle , Cell Line, Tumor , Cell Proliferation , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Gene Silencing , Humans , Pancreatic Neoplasms/metabolism , Promoter Regions, Genetic , Proto-Oncogene Proteins c-ets , Transcription, Genetic , Up-Regulation
5.
Carcinogenesis ; 38(8): 757-765, 2017 08 01.
Article in English | MEDLINE | ID: mdl-28430867

ABSTRACT

Although increased awareness leading to early detection and prevention, as well as advancements in treatment strategies, have resulted in superior clinical outcomes, African American women with breast cancer continue to have greater mortality rates, compared to Caucasian American counterparts. Moreover, African American women are more likely to have breast cancer at a younger age and be diagnosed with aggressive tumor sub-types. Such racial disparities can be attributed to socioeconomic differences, but it is increasingly being recognized that these disparities may indeed be due to certain genetic and other non-genetic biological differences. Tumor microenvironment, which provides a favorable niche for the growth of tumor cells, is comprised of several types of stromal cells and the various proteins secreted as a consequence of bi-directional tumor-stromal cross-talk. Emerging evidence suggests inherent biological differences in the tumor microenvironment of breast cancer patients from different racial backgrounds. Tumor microenvironment components, affected by the genetic make-up of the tumor cells as well as other non-tumor-associated factors, may also render patients more susceptible to the development of aggressive tumors and faster progression of disease resulting in early onset, thus adversely affecting patients' survival. This review provides an overview of breast cancer racial disparity and discusses the existence of race-associated differential tumor microenvironment and its underlying genetic and non-genetic causal factors. A better understanding of these aspects would help further research on effective cancer management and improved approaches for reducing the racial disparities gaps in breast cancer patients.


Subject(s)
Breast Neoplasms/epidemiology , Breast Neoplasms/genetics , Tumor Microenvironment/genetics , Black or African American/genetics , Breast Neoplasms/pathology , Female , Humans , Racial Groups/genetics , Risk Factors , Socioeconomic Factors , White People/genetics
6.
Cancer Lett ; 396: 21-29, 2017 06 28.
Article in English | MEDLINE | ID: mdl-28302531

ABSTRACT

Breast cancer (BC) continues to be the most frequently diagnosed cancer in American women, which disproportionately affects women of African-American (AA) descent. Previously, we reported greater serum levels of resistin in AA BC patients relative to Caucasian-American (CA) patients, and established its role in growth and aggressiveness of breast tumor cells. Here we have investigated the role of resistin in BC-chemoresistance. MDA-MB-231 and MDA-MB-468 BC cells of CA and AA origin, respectively, were incubated with resistin prior to doxorubicin treatment. Our data suggest that resistin conferred chemoresistance to both BC cell lines; however, the effect on AA cells was more profound. Furthermore, the resistin-induced doxorubicin-resistance was shown to occur due to suppression of apoptosis. Resistin treatment also affected the stemness of BC cells, as suggested by reduced cell surface expression of CD24, induced expression of CD44 and ALDH1, and increased capability of cells to form mammospheres. Mechanistic studies revealed that resistin-induced chemoresistance, apoptosis and stemness of BC cells were mediated through STAT3 activation. Taken together, our findings provide novel insight into the role of resistin in BC biology, and strengthen its role in racially disparate clinical outcomes.


Subject(s)
Breast Neoplasms/drug therapy , Doxorubicin/pharmacology , Health Status Disparities , Resistin/pharmacology , Antibiotics, Antineoplastic/pharmacology , Apoptosis/drug effects , Breast Neoplasms/metabolism , Breast Neoplasms/pathology , Drug Interactions , Drug Resistance, Neoplasm , Female , Humans , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Resistin/metabolism , Treatment Outcome
7.
Biochim Biophys Acta Rev Cancer ; 1868(1): 16-28, 2017 Aug.
Article in English | MEDLINE | ID: mdl-28108348

ABSTRACT

Despite efforts at various levels, racial health disparities still exist in cancer patients. These inequalities in incidence and/or clinical outcome can only be explained by a multitude of factors, with genetic basis being one of them. Several investigations have provided convincing evidence to support epigenetic regulation of cancer-associated genes, which results in the differential transcriptome and proteome, and may be linked to a pre-disposition of individuals of certain race/ethnicity to early or more aggressive cancers. Recent technological advancements and the ability to quickly analyze whole genome have aided in these efforts, and owing to their relatively easy detection, methylation events are much well-characterized, than the acetylation events, across human populations. The early trend of investigating a pre-determined set of genes for differential epigenetic regulation is paving way for more unbiased screening. This review summarizes our current understanding of the epigenetic events that have been tied to the racial differences in cancer incidence and mortality. A better understanding of the epigenetics of racial diversity holds promise for the design and execution of novel strategies targeting the human epigenome for reducing the disparity gaps.


Subject(s)
Epigenesis, Genetic/genetics , Neoplasms/genetics , Acetylation , Animals , DNA Methylation/genetics , Humans , Proteome/genetics , Transcriptome/genetics
8.
Front Biosci (Landmark Ed) ; 22(5): 772-782, 2017 01 01.
Article in English | MEDLINE | ID: mdl-27814645

ABSTRACT

Prostate cancer incidence and mortality rates are remarkably higher in African-American men as compared to their European-Americans counterparts. Despite these recognitions, precise causes underlying such prevalent racial disparities remain poorly understood. Although socioeconomic factors could account for such differences up to a certain extent, it is now being increasingly realized that such disparity has a molecular basis. Indeed, several differences, including genetic polymorphism, gene mutations, epigenetic modifications, miRNAs alterations, etc., have been reported in malignant prostate tissues from patients of diverse racial backgrounds. Here, we attempt to provide a molecular perspective on prostate cancer racial disparities by gathering available information on these associated factors and discussing their potential significance in disproportionate incidence and clinical outcomes.


Subject(s)
Black or African American/genetics , Prostatic Neoplasms/epidemiology , Prostatic Neoplasms/genetics , White People/genetics , Epigenesis, Genetic , Genetic Predisposition to Disease , Health Behavior , Humans , Incidence , Male , MicroRNAs/genetics , Mutation , Polymorphism, Genetic , Prostatic Neoplasms/etiology , Signal Transduction , Socioeconomic Factors , United States/epidemiology
9.
Cancer Lett ; 383(1): 53-61, 2016 12 01.
Article in English | MEDLINE | ID: mdl-27693632

ABSTRACT

Sunscreen formulations containing UVB filters, such as Zinc-oxide (ZnO) and titanium-dioxide (TiO2) nanoparticles (NPs) have been developed to limit the exposure of human skin to UV-radiations. Unfortunately, these UVB protective agents have failed in controlling the skin cancer incidence. We recently demonstrated that silver nanoparticles (Ag-NPs) could serve as novel protective agents against UVB-radiations. Here our goal was to perform comparative analysis of direct and indirect UVB-protection efficacy of ZnO-, TiO2- and Ag-NPs. Sun-protection-factor calculated based on their UVB-reflective/absorption abilities was the highest for TiO2-NPs followed by Ag- and ZnO-NPs. This was further confirmed by studying indirect protection of UVB radiation-induced death of HaCaT cells. However, only Ag-NPs were active in protecting HaCaT cells against direct UVB-induced DNA-damage by repairing bulky-DNA lesions through nucleotide-excision-repair mechanism. Moreover, Ag-NPs were also effective in protecting HaCaT cells from UVB-induced oxidative DNA damage by enhancing SOD/CAT/GPx activity. In contrast, ZnO- and TiO2-NPs not only failed in providing any direct protection from DNA-damage, but rather enhanced oxidative DNA-damage by increasing ROS production. Together, these findings raise concerns about safety of ZnO- and TiO2-NPs and establish superior protective efficacy of Ag-NPs.


Subject(s)
Anticarcinogenic Agents/pharmacology , DNA Damage/drug effects , Keratinocytes/drug effects , Keratinocytes/radiation effects , Metal Nanoparticles , Silver Compounds/pharmacology , Skin Neoplasms/prevention & control , Sunburn/drug therapy , Sunscreening Agents/pharmacology , Titanium/pharmacology , Ultraviolet Rays/adverse effects , Zinc Oxide/pharmacology , Anticarcinogenic Agents/toxicity , Antioxidants/pharmacology , Catalase/metabolism , Cell Death/drug effects , Cell Line, Tumor , DNA Repair/drug effects , Dose-Response Relationship, Drug , Glutathione Peroxidase/metabolism , Humans , Keratinocytes/metabolism , Keratinocytes/pathology , Oxidative Stress/drug effects , Reactive Oxygen Species/metabolism , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Skin Neoplasms/pathology , Sunburn/genetics , Sunburn/metabolism , Sunburn/pathology , Sunscreening Agents/toxicity , Superoxide Dismutase/metabolism , Titanium/toxicity , Zinc Oxide/toxicity
10.
Carcinogenesis ; 37(11): 1052-1061, 2016 11 01.
Article in English | MEDLINE | ID: mdl-27609457

ABSTRACT

The poor clinical outcome of pancreatic cancer (PC) is largely attributed to its aggressive nature and refractoriness to currently available therapeutic modalities. We previously reported antitumor efficacy of honokiol (HNK), a phytochemical isolated from various parts of Magnolia plant, against PC cells in short-term in vitro growth assays. Here, we report that HNK reduces plating efficiency and anchorage-independent growth of PC cells and suppresses their migration and invasiveness. Furthermore, significant inhibition of pancreatic tumor growth by HNK is observed in orthotopic mouse model along with complete-blockage of distant metastases. Histological examination suggests reduced desmoplasia in tumors from HNK-treated mice, later confirmed by immunohistochemical analyses of myofibroblast and extracellular matrix marker proteins (α-SMA and collagen I, respectively). At the molecular level, HNK treatment leads to decreased expression of sonic hedgehog (SHH) and CXCR4, two established mediators of bidirectional tumor-stromal cross-talk, both in vitro and in vivo . We also show that the conditioned media (CM) from HNK-treated PC cells have little growth-inducing effect on pancreatic stellate cells (PSCs) that could be regained by the addition of exogenous recombinant SHH. Moreover, pretreatment of CM of vehicle-treated PC cells with SHH-neutralizing antibody abolishes their growth-inducing potential on PSCs. Likewise, HNK-treated PC cells respond poorly to CM from PSCs due to decreased CXCR4 expression. Lastly, we show that the transfection of PC cells with constitutively active IKKß mutant reverses the suppressive effect of HNK on nuclear factor-kappaB activation and partially restores CXCR4 and SHH expression. Taken together, these findings suggest that HNK interferes with tumor-stromal cross-talk via downregulation of CXCR4 and SHH and decreases pancreatic tumor growth and metastasis.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Biphenyl Compounds/pharmacology , Lignans/pharmacology , Liver Neoplasms, Experimental/prevention & control , Lung Neoplasms/prevention & control , Pancreatic Neoplasms/drug therapy , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Biphenyl Compounds/therapeutic use , Cell Communication , Cell Line, Tumor , Down-Regulation , Female , Gene Expression/drug effects , Hedgehog Proteins/genetics , Hedgehog Proteins/metabolism , Humans , Lignans/therapeutic use , Liver Neoplasms, Experimental/secondary , Lung Neoplasms/secondary , Mice , Pancreatic Neoplasms/pathology , Receptors, CXCR4/genetics , Receptors, CXCR4/metabolism , Tumor Microenvironment , Xenograft Model Antitumor Assays
11.
J Biol Chem ; 291(31): 16263-70, 2016 07 29.
Article in English | MEDLINE | ID: mdl-27246849

ABSTRACT

Extensive desmoplasia is a prominent pathological characteristic of pancreatic cancer (PC) that not only impacts tumor development, but therapeutic outcome as well. Recently, we demonstrated a novel role of MYB, an oncogenic transcription factor, in PC growth and metastasis. Here we studied its effect on pancreatic tumor histopathology and associated molecular and biological mechanisms. Tumor-xenografts derived from orthotopic-inoculation of MYB-overexpressing PC cells exhibited far-greater desmoplasia in histological analyses compared with those derived from MYB-silenced PC cells. These findings were further confirmed by immunostaining of tumor-xenograft sections with collagen-I, fibronectin (major extracellular-matrix proteins), and α-SMA (well-characterized marker of myofibroblasts or activated pancreatic stellate cells (PSCs)). Likewise, MYB-overexpressing PC cells provided significantly greater growth benefit to PSCs in a co-culture system as compared with the MYB-silenced cells. Interrogation of deep-sequencing data from MYB-overexpressing versus -silenced PC cells identified Sonic-hedgehog (SHH) and Adrenomedullin (ADM) as two differentially-expressed genes among others, which encode for secretory ligands involved in tumor-stromal cross-talk. In-silico analyses predicted putative MYB-binding sites in SHH and ADM promoters, which was later confirmed by chromatin-immunoprecipitation. A cooperative role of SHH and ADM in growth promotion of PSCs was confirmed in co-culture by using their specific-inhibitors and exogenous recombinant-proteins. Importantly, while SHH acted exclusively in a paracrine fashion on PSCs and influenced the growth of PC cells only indirectly, ADM could directly impact the growth of both PC cells and PSCs. In summary, we identified MYB as novel regulator of pancreatic tumor desmoplasia, which is suggestive of its diverse roles in PC pathobiology.


Subject(s)
Adrenomedullin/biosynthesis , Gene Expression Regulation, Neoplastic , Hedgehog Proteins/biosynthesis , Oncogene Proteins v-myb/metabolism , Pancreatic Neoplasms/metabolism , Paracrine Communication , Response Elements , Transcription, Genetic , Up-Regulation , Adrenomedullin/genetics , Animals , Cell Line, Tumor , Hedgehog Proteins/genetics , Heterografts , Humans , Mice , Neoplasm Transplantation , Oncogene Proteins v-myb/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Pancreatic Stellate Cells/metabolism , Pancreatic Stellate Cells/pathology
12.
Cancer Lett ; 370(2): 260-7, 2016 Jan 28.
Article in English | MEDLINE | ID: mdl-26546043

ABSTRACT

Pancreatic cancer (PC) remains a highly lethal malignancy due to its unusual chemoresistance and high aggressiveness. A subpopulation of pancreatic tumor cells, known as cancer stem cells (CSCs), is considered responsible not only for tumor-maintenance, but also for its widespread metastasis and therapeutic failure. Here we investigated the role of p-21 activated kinase 4 (PAK4) in driving PC stemness properties. Our data demonstrate that triple-positive (CD24(+)/CD44(+)/EpCAM(+)) subpopulation of pancreatic CSCs exhibits greater level of PAK4 as compared to triple-negative (CD24(-)/CD44(-)/EpCAM(-)) cells. Moreover, PAK4 silencing in PC cells leads to diminished fraction of CD24, CD44, and EpCAM positive cells. Furthermore, we show that PAK4-silenced PC cells exhibit decreased sphere-forming ability and increased chemosensitivity to gemcitabine toxicity. PAK4 expression is also associated with enhanced levels of stemness-associated transcription factors (Oct4/Nanog/Sox2 and KLF4). Furthermore, our data show decreased nuclear accumulation and transcriptional activity of STAT3 in PAK4-silenced PC cells and restitution of its activity leads to restoration of stem cell phenotypes. Together, our findings deliver first experimental evidence for the involvement of PAK4 in PC stemness and support its clinical utility as a novel therapeutic target in PC.


Subject(s)
Neoplastic Stem Cells/chemistry , Pancreatic Neoplasms/pathology , STAT3 Transcription Factor/physiology , Signal Transduction/physiology , p21-Activated Kinases/physiology , Aldehyde Dehydrogenase 1 Family , Antigens, Neoplasm/analysis , CD24 Antigen/analysis , Cell Adhesion Molecules/analysis , Cell Line, Tumor , Epithelial Cell Adhesion Molecule , Humans , Hyaluronan Receptors/analysis , Isoenzymes/physiology , Kruppel-Like Factor 4 , Phenotype , Retinal Dehydrogenase/physiology
13.
Br J Cancer ; 113(12): 1694-703, 2015 Dec 22.
Article in English | MEDLINE | ID: mdl-26657649

ABSTRACT

BACKGROUND: MYB encodes for a transcription factor regulating the expression of a wide array of genes involved in cellular functions. It is reported to be amplified in a sub-set of pancreatic cancer (PC) cases; however, its pathobiological association has remained unclear thus far. METHODS: Expression of MYB and other cellular proteins was analysed by immunoblot or qRT-PCR analyses. MYB was stably overexpressed in non-expressing (BxPC3) and silenced in highly expressing (MiaPaCa and Panc1) PC cells. Effect on growth was analysed by automated cell counting at 24-h interval. Cell-cycle progression and apoptotic indices of PC cells with altered MYB expression were measured through flow cytometry upon staining with respective biomarkers. Cell motility/invasion was examined in a Boyden's chamber assay using non-coated or Matrigel-coated membranes. Effect on tumorigenicity and metastatic potential was examined by non-invasive imaging and through end-point measurements of luciferase-tagged MYB-altered PC implanted in the pancreas of nude mice. RESULTS: MYB was aberrantly expressed in all malignant cases of pancreas, whereas remained undetectable in normal pancreas. All the tested established PC cell lines except BxPC3 also exhibited MYB expression. Forced expression of MYB in BxPC3 cells promoted their growth, cell-cycle progression, survival and malignant behaviour, whereas its silencing in MiaPaCa and Panc1 cells produced converse effects. More importantly, ectopic MYB expression was sufficient to confer tumorigenic and metastatic capabilities to non-tumorigenic BxPC3 cells, while its silencing resulted in significant loss of the same in MYB-overexpressing cells as demonstrated in orthotopic mouse model. We also identified several MYB-regulated genes in PC cells that might potentially mediate its effect on tumour growth and metastasis. CONCLUSIONS: MYB is aberrantly overexpressed in PC cells and acts as a key determinant of pancreatic tumour growth and metastasis.


Subject(s)
Cell Division/genetics , Genes, myb , Neoplasm Metastasis/genetics , Pancreatic Neoplasms/pathology , Animals , Cell Cycle , Heterografts , Humans , Mice , Pancreatic Neoplasms/genetics
14.
BMC Cancer ; 15: 636, 2015 Sep 15.
Article in English | MEDLINE | ID: mdl-26373391

ABSTRACT

BACKGROUND: Ultraviolet (UV) radiation from sun, particularly its UVB component (290-320 nm), is considered the major etiological cause of skin cancer that impacts over 2 million lives in the United States alone. Recently, we reported that polydisperse colloidal suspension of silver nanoparticles (AgNPs) protected the human keratinocytes (HaCaT) against UVB-induced damage, thus indicating their potential for prevention of skin carcinogenesis. Here we sought out to investigate if size controlled the chemopreventive efficacy of AgNPs against UVB-induced DNA damage and apoptosis. METHODS: Percent cell viability was examined by WST-1 assay after treating the cells with various doses (1-10 µg/mL) of AgNPs of different sizes (10, 20, 40, 60 and 100 nm) for 12 and 24 h. For protection studies, cells were treated with AgNPs of different sizes at a uniform concentration of 1 µg/mL. After 3 h, cells were irradiated with UVB (40 mJ/cm(2)) and dot-blot analysis was performed to detect cyclobutane pyrimidine dimers (CPDs) as an indication of DNA damage. Apoptosis was analyzed by flow cytometry after staining the cells with 7-Amino-Actinomycin (7-AAD) and PE Annexin V. Immunoblot analysis was accomplished by processing the cells for protein extraction and Western blotting using specific antibodies against various proteins. RESULTS: The data show that the pretreatment of HaCaT cells with AgNPs in the size range of 10-40 nm were effective in protecting the skin cells from UVB radiation-induced DNA damage as validated by reduced amounts of CPDs, whereas no protection was observed with AgNPs of larger sizes (60 and 100 nm). Similarly, only smaller size AgNPs (10-40 nm) were effective in protecting the skin cells from UV radiation-induced apoptosis. At the molecular level, UVB -irradiation of HaCaT cells led to marked increase in expression of pro-apoptotic protein (Bax) and decrease in anti-apoptotic proteins (Bcl-2 and Bcl-xL), while it remained largely unaffected in skin cells pretreated with smaller size AgNPs (10-40 nm). CONCLUSIONS: Altogether, these findings suggest that size is a critical determinant of the UVB-protective efficacy of AgNPs in human keratinocytes.


Subject(s)
Keratinocytes/drug effects , Keratinocytes/radiation effects , Metal Nanoparticles , Protective Agents , Silver , Ultraviolet Rays , Apoptosis/drug effects , Apoptosis/radiation effects , Cell Line, Tumor , Cell Survival/drug effects , Cell Survival/radiation effects , DNA Damage/drug effects , DNA Damage/radiation effects , Humans , Metal Nanoparticles/administration & dosage , Metal Nanoparticles/chemistry , Particle Size , Protective Agents/administration & dosage , Protective Agents/chemistry
15.
Sci Rep ; 5: 13894, 2015 Sep 09.
Article in English | MEDLINE | ID: mdl-26349906

ABSTRACT

Epidemiological studies suggest ultraviolet B (UVB) component (290-320 nm) of sun light is the most prevalent etiologic factor for skin carcinogenesis--a disease accounting for more than two million new cases each year in the USA alone. Development of UVB-induced skin carcinoma is a multistep and complex process. The molecular events that occur during UVB-induced skin carcinogenesis are poorly understood largely due to the lack of an appropriate cellular model system. Therefore, to make a progress in this area, we have developed an in vitro model for UVB-induced skin cancer using immortalized human epidermal keratinocyte (HaCaT) cells through repetitive exposure to UVB radiation. We demonstrate that UVB-transformed HaCaT cells gain enhanced proliferation rate, apoptosis-resistance, and colony- and sphere-forming abilities in a progressive manner. Moreover, these cells exhibit increased aggressiveness with enhanced migration and invasive potential and mesenchymal phenotypes. Furthermore, these derived cells are able to form aggressive squamous cell carcinoma upon inoculation into the nude mice, while parental HaCaT cells remain non-tumorigenic. Together, these novel, UVB-transformed progression model cell lines can be very helpful in gaining valuable mechanistic insight into UVB-induced skin carcinogenesis, identification of novel molecular targets of diagnostic and therapeutic significance, and in vitro screening for novel preventive and therapeutic agents.


Subject(s)
Cell Transformation, Neoplastic/radiation effects , Skin Neoplasms/etiology , Skin Neoplasms/pathology , Ultraviolet Rays/adverse effects , Animals , Apoptosis/radiation effects , Cell Line, Tumor , Cell Movement/radiation effects , Cell Proliferation/radiation effects , Disease Models, Animal , Disease Progression , Epithelial-Mesenchymal Transition/radiation effects , Heterografts , Humans , In Vitro Techniques , Mice
16.
Oncotarget ; 6(36): 39140-50, 2015 Nov 17.
Article in English | MEDLINE | ID: mdl-25970774

ABSTRACT

Pancreatic tumor microenvironment (TME) is characterized by poor tumor-vasculature and extensive desmoplasia that together contribute to poor response to chemotherapy. It was recently shown that targeting of TME to inhibit desmoplasiatic reaction in a preclinical model resulted in increased microvessel-density and intratumoral drug concentration, leading to improved therapeutic response. This approach, however, failed to generate a favorable response in clinical trial. In that regard, we have previously demonstrated a role of gemcitabine-induced CXCR4 signaling as a counter-defense mechanism, which also promoted invasiveness of pancreatic cancer (PC) cells. Here, we investigated the effect of gemcitabine on endothelial cell phenotype. Gemcitabine-treatment of human-umbilical-vein-endothelial-cells (HUVECs) did not promote the growth of HUVECs; however, it was induced when treated with conditioned media from gemcitabine-treated (Gem-CM) PC cells due to increased cell-cycle progression and apoptotic-resistance. Moreover, treatment of HUVECs with Gem-CM resulted in capillary-like structure (CLS) formation and promoted their ability to migrate and invade through extracellular-matrix. Gemcitabine-treatment of PC cells induced expression of various growth factors/cytokines, including IL-8, which exhibited greatest upregulation. Further, IL-8 depletion in Gem-CM diminished its potency to promote angiogenic phenotypes. Together, these findings suggest an indirect effect of gemcitabine on angiogenesis, which, in light of our previous observations, may hold important clinical significance.


Subject(s)
Antimetabolites, Antineoplastic/pharmacology , Deoxycytidine/analogs & derivatives , Endothelial Cells/drug effects , Pancreatic Neoplasms/blood supply , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Deoxycytidine/pharmacology , Endothelial Cells/metabolism , Endothelial Cells/pathology , Human Umbilical Vein Endothelial Cells , Humans , Neovascularization, Pathologic/metabolism , Neovascularization, Pathologic/pathology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , Signal Transduction , Survival Analysis , Tumor Microenvironment , Gemcitabine
17.
Oncotarget ; 6(13): 11231-41, 2015 May 10.
Article in English | MEDLINE | ID: mdl-25868978

ABSTRACT

African-American (AA) women with breast cancer (BC) are diagnosed with more aggressive disease, have higher risk of recurrence and poorer prognosis as compared to Caucasian American (CA) women. Therefore, it is imperative to define the factors associated with such disparities to reduce the unequal burden of cancer. Emerging data suggest that inherent differences exist in the tumor microenvironment of AA and CA BC patients, however, its molecular bases and functional impact have remained poorly understood. Here, we conducted cytokine profiling in serum samples from AA and CA BC patients and identified resistin and IL-6 to be the most differentially-expressed cytokines with relative greater expression in AA patients. Resistin and IL-6 exhibited positive correlation in serum levels and treatment of BC cells with resistin led to enhanced production of IL-6. Moreover, resistin also enhanced the expression and phosphorylation of STAT3, and treatment of BC cells with IL-6-neutralizing antibody prior to resistin stimulation abolished STAT3 phosphorylation. In addition, resistin promoted growth and aggressiveness of BC cells, and these effects were mediated through STAT3 activation. Together, these findings suggest a crucial role of resistin, IL-6 and STAT3 in BC racial disparity.


Subject(s)
Biomarkers, Tumor/blood , Black or African American , Breast Neoplasms/blood , Cell Proliferation , Health Status Disparities , Interleukin-6/blood , Resistin/blood , STAT3 Transcription Factor/metabolism , White People , Black or African American/genetics , Antibodies, Neutralizing/pharmacology , Antineoplastic Agents/pharmacology , Biomarkers, Tumor/genetics , Breast Neoplasms/drug therapy , Breast Neoplasms/ethnology , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Cell Line, Tumor , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Female , Genetic Predisposition to Disease/ethnology , Humans , Interleukin-6/antagonists & inhibitors , Interleukin-6/genetics , Neoplasm Invasiveness , Phenotype , Phosphorylation , RNA Interference , Resistin/pharmacology , STAT3 Transcription Factor/genetics , Signal Transduction , Time Factors , Transfection , Tumor Microenvironment , White People/genetics
18.
Nanomedicine ; 11(5): 1265-75, 2015 Jul.
Article in English | MEDLINE | ID: mdl-25804413

ABSTRACT

Ultraviolet (UV)-B radiation from the sun is an established etiological cause of skin cancer, which afflicts more than a million lives each year in the United States alone. Here, we tested the chemopreventive efficacy of silver-nanoparticles (AgNPs) against UVB-irradiation-induced DNA damage and apoptosis in human immortalized keratinocytes (HaCaT). AgNPs were synthesized by reduction-chemistry and characterized for their physicochemical properties. AgNPs were well tolerated by HaCaT cells and their pretreatment protected them from UVB-irradiation-induced apoptosis along with significant reduction in cyclobutane-pyrimidine-dimer formation. Moreover, AgNPs pre-treatment led to G1-phase cell-cycle arrest in UVB-irradiated HaCaT cells. AgNPs were efficiently internalized in UVB-irradiated cells and localized into cytoplasmic and nuclear compartments. Furthermore, we observed an altered expression of various genes involved in cell-cycle, apoptosis and nucleotide-excision repair in HaCaT cells treated with AgNPs prior to UVB-irradiation. Together, these findings provide support for potential utility of AgNPs as novel chemopreventive agents against UVB-irradiation-induced skin carcinogenesis. FROM THE CLINICAL EDITOR: Excessive exposure to the sun is known to increase the risk of skin cancer due to DNA damage. In this work, the authors tested the use of silver nanoparticles as protective agents against ultraviolet radiation. The positive results may open a door for the use of silver nanoparticle as novel agents in the future.


Subject(s)
Anticarcinogenic Agents/pharmacology , Apoptosis/drug effects , Keratinocytes/drug effects , Keratinocytes/radiation effects , Metal Nanoparticles , Silver/pharmacology , Anticarcinogenic Agents/chemistry , Cell Line , DNA Damage/drug effects , DNA Repair , Humans , Keratinocytes/pathology , Metal Nanoparticles/chemistry , Metal Nanoparticles/ultrastructure , Silver/chemistry , Skin Neoplasms/prevention & control , Ultraviolet Rays
19.
Oncotarget ; 5(22): 11490-500, 2014 Nov 30.
Article in English | MEDLINE | ID: mdl-25359780

ABSTRACT

Emerging data highlight the significance of chemokine (C-X-C motif) ligand 12/chemokine (C-X-C motif) receptor 4 (CXCL12/CXCR4) signaling axis in the chemoresistance of several malignancies, including prostate cancer (PCa); however, underlying mechanisms remain largely elusive. Here, we demonstrate that CXCL12 treatment rescues the PCa cells from docetaxel (DTX)-induced toxicity by overriding its effect on cell cycle (G2/M phase arrest). We further demonstrate that the chemoprotective effect of CXCL12 is abolished upon pharmacological inhibition or RNA interference-mediated silencing of CXCR4. Moreover, microtubule stabilization caused by DTX is suppressed in CXCL12-stimulated PCa cells as revealed by immunofluorescence and immunoblot analyses. The effect of CXCL12 on microtubule stabilization is abrogated when PCa cells are pre-treated with a CXCR4 antagonist. In additional studies, we show that the chemoprotective action of CXCL12/CXCR4 signaling is mediated by p21-activated kinase 4 (PAK4)-dependent activation of Lim domain kinase 1 (LIMK1), and inhibition of either PAK4 or LIMK1 leads to re-sensitization of PCa cells to DTX-induced tubulin polymerization and cellular toxicity even in the presence of CXCL12. Altogether, our findings uncover a novel mechanism underlying CXCL12/CXCR4 signaling-induced PCa chemoresistance and suggest that targeting of this signaling axis or its downstream effector pathway could lead to therapeutic enhancement of DTX.


Subject(s)
Chemokine CXCL12/metabolism , Lim Kinases/metabolism , Microtubules/metabolism , Prostatic Neoplasms/metabolism , Receptors, CXCR4/metabolism , Taxoids/chemistry , p21-Activated Kinases/metabolism , Anticarcinogenic Agents/chemistry , Cell Cycle , Cell Division , Cell Line, Tumor , Docetaxel , Drug Resistance, Neoplasm , G2 Phase , Gene Expression Regulation, Neoplastic , Humans , Male , Microscopy, Fluorescence , Phosphorylation , RNA Interference , Signal Transduction
20.
Oncotarget ; 5(18): 8778-89, 2014 Sep 30.
Article in English | MEDLINE | ID: mdl-25238288

ABSTRACT

Identification of novel molecular targets and understanding the mechanisms underlying the aggressive nature of pancreatic cancer (PC) remain prime focus areas of research. Here, we investigated the expression and pathobiological significance of p21-activated kinase 4 (PAK4), a gene that was earlier shown to be amplified in a sub-set of PC. Our data demonstrate PAK4 overexpression in PC tissues and cell lines with little or no expression in the normal pancreas. PAK4 silencing in two PC cell lines, MiaPaCa and T3M4, by RNA interference causes suppression of growth and clonogenic ability due to decreased cell cycle progression and apoptosis-resistance. PAK4-silenced PC cells exhibit altered expression of proliferation- and survival-associated proteins. Moreover, we observe decreased nuclear accumulation and transcriptional activity of NF-κB in PAK4-silenced PC cells associated with stabilization of its inhibitory protein, IκBα. Transfection of PAK4-silenced PC cells with constitutively-active mutant of IKKß, an upstream kinase of IκBα, leads to restoration of NF-κB activity and PC cell growth. Furthermore, we show that PAK4-induced NF-κB activity is mediated through activation and concerted action of ERK and Akt kinases. Together, these findings suggest that PAK4 is a regulator of NF-κB pathway in PC cells and can serve as a novel target for therapy.


Subject(s)
Cell Proliferation , Extracellular Signal-Regulated MAP Kinases/metabolism , NF-kappa B/metabolism , Pancreatic Neoplasms/enzymology , Proto-Oncogene Proteins c-akt/metabolism , p21-Activated Kinases/metabolism , Apoptosis , Cell Cycle , Cell Line, Tumor , Cell Proliferation/drug effects , Cell Survival , Drug Resistance, Neoplasm , Enzyme Activation , Extracellular Signal-Regulated MAP Kinases/antagonists & inhibitors , Gene Expression Regulation, Neoplastic , Humans , NF-kappa B/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/pathology , Protein Kinase Inhibitors/pharmacology , Proto-Oncogene Proteins c-akt/antagonists & inhibitors , RNA Interference , Signal Transduction , Time Factors , Transfection , p21-Activated Kinases/genetics
SELECTION OF CITATIONS
SEARCH DETAIL
...