Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 43
Filter
1.
Environ Sci Pollut Res Int ; 30(36): 84999-85045, 2023 Aug.
Article in English | MEDLINE | ID: mdl-37400699

ABSTRACT

The release of contaminants of environmental concern including heavy metals and metalloids, and contaminants of emerging concern including organic micropollutants from processing industries, pharmaceuticals, personal care, and anthropogenic sources, is a growing threat worldwide. Mitigating inorganic and organic contaminants, which can be coined as contaminants of environmental and emerging concern (CEECs), is a big challenge as traditional physicochemical processes are not economically viable for managing mixed contaminants of low concentrations. As a result, low-cost materials must be designed to provide high CEEC removal efficiency. One of the environmentally viable and energy-efficient approaches is biosorption, which involves using biomass or biopolymers isolated from plants or animals to decontaminate heavy metals in contaminated environments using inherent biological mechanisms. Among chemical constituents in plant biomass, cellulose, lignin, hemicellulose, proteins, polysaccharides, phenolic compounds, and animal biomass include polysaccharides and other compounds to bind heavy metals covalently and non-covalently. These functional groups include carboxyl, hydroxyl, carbonyl, amide, amine, and sulfhydryl. Cation-exchange capacities of these bioadsorbents can be improved by applying chemical modifications. The relevance of chemical constituents and bioactives in biosorbents derived from agricultural production such as food and fodder crops, bioenergy and cash crops, fruit and vegetable crops, medicinal and aromatic plants, plantation trees, aquatic and terrestrial weeds, and animal production such as dairy, goatery, poultry, duckery, and fisheries is highlighted in this comprehensive review for sequestering and bioremediation of CEECs, including as many as ten different heavy metals and metalloids co-contaminated with other organic micropollutants in circular bioresource utilization and one-health concepts.


Subject(s)
Metalloids , Metals, Heavy , Animals , Biodegradation, Environmental , Fisheries , Metals, Heavy/metabolism , Agriculture , Plants/metabolism
2.
Sci Rep ; 12(1): 13691, 2022 Aug 11.
Article in English | MEDLINE | ID: mdl-35953697

ABSTRACT

Localization in underwater wireless sensor network (UWSN) faces an imminent threat when the triangulating anchor node starts to malfunction. Traditional geometric approaches are insufficient to cope with the survivability of UWSN topology. To address these issues, this paper presents a symplectic geometry for identification of the malicious anchor node. Consequently, a geodesic search algorithm (GSA) based Target localization is proposed which reduces the positioning error by exploiting the phase-space constancy of the underwater acoustic sensor network topology to effectively triangulate the target node despite its mobility. First, a malicious anchor node model is presented. The node movement is expressed in the form of "ripple region". GSA is then proposed which effectively frees the node metastasis from anchor node geometry, thereby making the underwater system more survivable and resilient. Simulation results evaluate the survivability of the geodesic formalism in terms of the reduced penalty incurred by node movement, as well as the reduced impact of anchor node malfunction. An improvement of 13.46% and 9.26% reveals the utility of the geodesic technique in aquamarine sensor deployments, which would be beneficial in underwater resource exploration and defense planning.

3.
J Fish Biol ; 101(1): 249-261, 2022 Jul.
Article in English | MEDLINE | ID: mdl-35593382

ABSTRACT

This study evaluated the effect of different potassium supplementation dosages on the physiological responses of Pangasianodon hypophthalmus reared in an aquaponic system with Spinacia oleracea L. for 60 days. The system comprised of a rectangular fish tank of 168 l capacity (water volume = 100 l) with nutrient film technique (NFT)-based hydroponic component with fish to plant ratio of 2.8 kg m-3 : 28 plants m-2 in all the treatments. The osmoregulatory and stress parameters of P. hypophthalmus at four different potassium dosages of T1 (90 mg l-1 ), T2 (120 mg l-1 ), T3 (150 mg l-1 ) and T4 (180 mg l-1 ) were compared with C (control, 0 mg l-1 ) to examine the potassium level to be applied to aquaponics. The water quality parameters and fish production were found to have no adverse impact due to potassium supplementation. The spinach yield during two harvests, i.e., before and after potassium supplementation, revealed that the yield was significantly higher (P < 0.05) after supplementation with the highest yield in T3 and T4. The osmoregulatory parameters such as plasma osmolality, Na+ , K+ ATPase activity in gill and plasma ionic profile (Cl- , Ca2+ and Na+ ) showed an insignificant variation (P > 0.05) between control and treatments except for higher plasma potassium concentration (1.98 ± 0.19 mmol l-1 ) in T4. The stress and antioxidant enzyme analysis exhibited significantly higher plasma glucose and superoxide dismutase (SOD) activity in gill and liver in T4, whereas cortisol and catalase showed an insignificant difference (P > 0.05). The experimental findings demonstrated that the potassium dosage up to 150 mg l-1 could be suggested as optimum for P. hypophthalmus and spinach aquaponics without impairing the health and oxidative status of P. hypophthalmus.


Subject(s)
Catfishes , Spinacia oleracea , Animals , Catfishes/physiology , Dietary Supplements , Gills , Potassium/pharmacology
4.
Cells ; 9(1)2020 01 14.
Article in English | MEDLINE | ID: mdl-31947623

ABSTRACT

Androgen receptor (AR) signaling remains crucial in castration-resistant prostate cancer (CRPC). Since it is also essential in immune cells, we studied whether the expression of AR full-length (ARFL) and its splicing variant ARV7 in peripheral blood mononuclear cells (PBMC) predicts systemic treatment response in mCRPC in comparison with circulating-tumor cells (CTC). We measured ARFL and ARV7 mRNA in PBMC and CTC from patients prior to receiving abiraterone (AA), enzalutamide (E), or taxanes by a pre-amplification plus quantitative reverse-transcription PCR. They were also tested in LNCaP-ARV7-transfected and in 22RV1 docetaxel-resistant (22RV1DR) cells. We studied 171 PBMC from 136 patients and from 24 non-cancer controls, and 47 CTC from 22 patients. High PBMC ARV7 levels correlated with worse AA/E and better taxane response. In taxane-treated patients high PBMC ARFL also correlated with longer progression-free survival (PFS). High ARV7 and ARFL expression were independently associated with better biochemical-PFS. Conversely, high CTC ARV7 and ARFL correlated with shorter radiological-PFS and overall survival, respectively. High ARV7 in 22RV1DR and LNCaP-ARV7 cells correlated with taxane resistance. In conclusion, ARFL and ARV7 at PBMC or CTC have a different predictive role in the taxane response, suggesting a potential influence of the AR pathway from PBMC in such response modulation.


Subject(s)
Leukocytes, Mononuclear/metabolism , Neoplastic Cells, Circulating/metabolism , Prostatic Neoplasms, Castration-Resistant/metabolism , Receptors, Androgen/genetics , Adult , Aged , Cell Line, Tumor , Female , Genetic Variation/genetics , Humans , Male , Middle Aged , Neoplastic Cells, Circulating/pathology , PC-3 Cells , Prostatic Neoplasms, Castration-Resistant/pathology , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Androgen/metabolism , Young Adult
5.
Mol Cancer Ther ; 15(12): 2863-2874, 2016 12.
Article in English | MEDLINE | ID: mdl-27765854

ABSTRACT

Although treatment of prostate cancer has improved over the past several years, taxanes, such as cabazitaxel, remain the only form of effective chemotherapy that improves survival in patients with metastatic castration-resistant prostate cancer. However, the effectiveness of this class of drugs has been associated with various side effects and drug resistance. We previously reported that fisetin, a hydroxyflavone, is a microtubule-stabilizing agent and inhibits prostate cancer cell proliferation, migration, and invasion and suggested its use as an adjuvant for treatment of prostate and other cancer types. In this study, we investigated the effect of fisetin in combination with cabazitaxel with the objective to achieve maximum therapeutic benefit, reduce dose and toxicity, and minimize or delay the induction of drug resistance and metastasis. Our data show for the first time that a combination of fisetin (20 µmol/L) enhances cabazitaxel (5 nmol/L) and synergistically reduces 22Rν1, PC-3M-luc-6, and C4-2 cell viability and metastatic properties with minimal adverse effects on normal prostate epithelial cells. In addition, the combination of fisetin with cabazitaxel was associated with inhibition of proliferation and enhancement of apoptosis. Furthermore, combination treatment resulted in the inhibition of tumor growth, invasion, and metastasis when assessed in two in vivo xenograft mouse models. These results provide evidence that fisetin may have therapeutic benefit for patients with advanced prostate cancer through enhancing the efficacy of cabazitaxel under both androgen-dependent and androgen-independent conditions. This study underscores the benefit of the combination of fisetin with cabazitaxel for the treatment of advanced and resistant prostate cancer and possibly other cancer types. Mol Cancer Ther; 15(12); 2863-74. ©2016 AACR.


Subject(s)
Antineoplastic Agents/pharmacology , Flavonoids/pharmacology , Taxoids/pharmacology , Animals , Apoptosis/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Disease Models, Animal , Drug Synergism , Drug Therapy, Combination , Flavonols , Humans , Male , Mice , Mice, Nude , Neoplastic Stem Cells/drug effects , Neoplastic Stem Cells/metabolism , Prostatic Neoplasms/diagnosis , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Treatment Outcome , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
6.
Oncotarget ; 7(51): 84924-84937, 2016 Dec 20.
Article in English | MEDLINE | ID: mdl-27793049

ABSTRACT

Cutaneous squamous cell carcinoma (cSCC) is the most common type of non-melanoma skin cancer that can metastasize. The major etiological factor associated with cSCC is Ultraviolet radiation (UVR) with a limited understanding of its molecular mechanism. It was hypothesized that there is a direct effect of UVR on modulation of microRNAs (miRNAs), a novel class of short noncoding RNAs which affects translation and stability of mRNAs. To test the hypothesis, the dorsal skin of the SKH1 mice (6-7 week old) was exposed to acute and chronic doses of UVR. In miRNA array profiling, we found differential expression (log fold change>1) of miR-25-5p between untreated and acute UVR treated (4kJ/m2) SKH1 mice skin. However, differential expression (>1 log fold) of miR-144-3p, miR-33-5p, miR-32-5p, miR-1983, miR-136-5p, miR-142-3p, miR-376a-3p, miR-142-5p, miR-3968, and miR-29b-3p was observed between untreated and chronically UVR treated mice skin. Differentially expressed selected miRNAs (miR-32-5p, miR-33-5p, miR-144-3p, and miR-376a-3p) were further validated in real time PCR using miRNA specific primers. Web based data mining, for the prediction of potential miRNA associated gene pathways in miRBase database revealed a link with important pathways (PI3K-Akt, MAPK, Wnt, transcriptional misregulation, and other oncogenic pathway) associated with cSCC. Furthermore, findings of PI3K-Akt pathway genes affected due to chronic UVR were confirmed using cDNA array.


Subject(s)
Carcinoma, Squamous Cell/genetics , Dermatology , MicroRNAs/genetics , Skin Neoplasms/genetics , Ultraviolet Rays , Animals , Cells, Cultured , Disease Models, Animal , Extracellular Signal-Regulated MAP Kinases/metabolism , Female , Gene Expression Profiling , Gene Expression Regulation/drug effects , Humans , Mice , Mice, Hairless , MicroRNAs/radiation effects , Organ Culture Techniques , Phosphatidylinositol 3-Kinases/metabolism , Research Design , Signal Transduction/genetics , Ultraviolet Rays/adverse effects , Wnt Proteins/metabolism
7.
J Photochem Photobiol B ; 162: 266-272, 2016 Sep.
Article in English | MEDLINE | ID: mdl-27394009

ABSTRACT

In the present work, monoclinic bismuth vanadate (m-BiVO4) nanostructures have been synthesized via simple hydrothermal method and employed for visible light driven antimicrobial and photocatalytic activity. Morphology (octahedral) and size (200-300nm) of the m-BiVO4 are studied using transmission electron microscopy (TEM). The crystal structure of m-BiVO4 (monoclinic scheelite structure) is confirmed by high resolution-TEM (HRTEM) and X-ray diffraction (XRD) studies. The band gap of m-BiVO4 was estimated to be ca. 2.42eV through Kubelka-Munk function F(R∞) using diffuse reflectance spectroscopy (DRS). Antimicrobial action of m-BiVO4 is anticipated by (i) shake flask method, (ii) MTT [3-(4,5-Dimethylthiazol-2-Yl)-2,5-Diphenyltetrazolium Bromide] assay for cytotoxicity. SEM analysis has been carried on Escherichia coli (E.coli) before and after treatment with nanostructure materials to reveal the mechanism underlying the antimicrobial action. Antimicrobial activity is studied as a function of m-BiVO4 concentration viz. 20, 40, 60 and 80ppm. The bacterial growth is decreased 80% to 96%, with the increase in m-BiVO4 concentration from 20ppm to 80ppm, respectively, in 2h. Photocatalytic activity and rate kinetics of m-BiVO4 nanostructures have been studied as a function of time on methylene blue (MB) dye degradation which is one of the waste products of textile industries and responsible for water pollution.


Subject(s)
Bismuth/chemistry , Bismuth/pharmacology , Light , Nanoparticles , Nanotechnology , Photochemical Processes , Temperature , Vanadates/chemistry , Vanadates/pharmacology , Anti-Bacterial Agents/chemical synthesis , Anti-Bacterial Agents/chemistry , Anti-Bacterial Agents/pharmacology , Chemistry Techniques, Synthetic , Escherichia coli/cytology , Escherichia coli/drug effects , Escherichia coli/radiation effects , Vanadates/chemical synthesis , Water Purification
8.
Oncotarget ; 7(22): 33069-80, 2016 May 31.
Article in English | MEDLINE | ID: mdl-27102301

ABSTRACT

PKCε is a transforming oncogene and a predictive biomarker of various human cancers. However, a precise in vivo link of PKCε to cancer induction, progression and metastasis remain undefined. To achieve these goals, we generated tissue specific conditional PKCε knockout mice (PKCε-CKO) using cre-lox technology. Homozygous PKCε(LoxP/LoxP) mice have normal body weight and phenotype. To determine what effect loss of PKCε would have on the prostate, the PKCε(LoxP/LoxP) mice were bred to probasin cre (PB-Cre4+) mice which express cre specifically in the prostate epithelium of postnatal mice. Western blot and immunohistochemical analyses showed reduced levels of PKCε specifically in the prostate of PKCε-CKO mice. Histopathological analyses of prostate from both PKCε(LoxP/LoxP) and prostate PKCε-CKO mice showed normal pathology. To determine the functional impact of prostate specific deletion of PKCε on prostate tumor growth, we performed an orthotopic xenograft study. Transgenic adenocarcinoma of the mouse prostate (TRAMP) cells (TRAMPC1, 2×106) were implanted in the prostate of PKCε-CKO mice. Mice were sacrificed at 6th week post-implantation. Results demonstrated a significant (P<0.05) decrease in the growth of TRAMPC1 cells-derived xenograft tumors in PKCε-CKO mice compared to wild type. To determine a link of PKCε to ultraviolet radiation (UVR) exposure-induced epidermal Stat3 phosphorylation, PKCε(LoxP/LoxP) mice were bred to tamoxifen-inducible K14 Cre mice. PKCε deletion in the epidermis resulted in inhibition of UVR-induced Stat3 phosphorylation. In summary, our novel PKCε(LoxP/LoxP) mice will be useful for defining the link of PKCε to various cancers in specific organ, tissue, or cells.


Subject(s)
Prostatic Neoplasms/genetics , Protein Kinase C-epsilon/metabolism , Animals , Disease Progression , Female , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Mice, Transgenic , Prostatic Neoplasms/pathology , Protein Kinase C-epsilon/deficiency , Protein Kinase C-epsilon/genetics
9.
Oncotarget ; 7(14): 17945-56, 2016 Apr 05.
Article in English | MEDLINE | ID: mdl-26918454

ABSTRACT

Chronic exposure to ultraviolet radiation (UVR) is linked to the development of cutaneous squamous cell carcinoma (SCC), a non-melanoma form of skin cancer that can metastasize. Tumor necrosis factor-alpha (TNFα), a pro-inflammatory cytokine, is linked to UVR-induced development of SCC. To find clues about the mechanisms by which TNFα may promote UVR-induced development of SCC, we investigated changes in the expression profiling of microRNAs (miRNA), a novel class of short noncoding RNAs, which affects translation and stability of mRNAs. In this experiment, TNFα knockout (TNFα KO) mice and their wild type (WT) littermates were exposed to acute UVR (2.0 kJ/m2) and the expression profiling of epidermal miRNA was determined 4hr post UVR exposure. TNFα deletion in untreated WT mice resulted in differential expression (log fold change>1) of seventeen miRNA. UVR exposure in WT mice induced differential expression of 22 miRNA. However, UVR exposure in TNFα KO mice altered only two miRNAs. Four miRNA, were differentially expressed between WT+UVR and TNFα KO+UVR groups. Differentially expressed selected miRNAs were further validated using real time PCR. Few of the differentially expressed miRNAs (miR-31-5p, miR-196a-5p, miR-127-3p, miR-206-3p, miR-411-5p, miR-709, and miR-322-5p) were also observed in UVR-induced SCC. Finally, bio-informatics analysis using DIANA, MIRANDA, Target Scan, and miRDB algorithms revealed a link with major UVR-induced pathways (MAPK, PI3K-Akt, transcriptional mis-regulation, Wnt, and TGF-beta).


Subject(s)
Carcinoma, Squamous Cell/etiology , Epidermis/metabolism , MicroRNAs/biosynthesis , Radiation Injuries, Experimental/etiology , Radiation Injuries, Experimental/metabolism , Skin Neoplasms/etiology , Tumor Necrosis Factor-alpha/biosynthesis , Ultraviolet Rays/adverse effects , Animals , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Epidermis/radiation effects , Mice , Mice, Knockout , MicroRNAs/genetics , Radiation Injuries, Experimental/genetics , Skin Neoplasms/genetics , Skin Neoplasms/metabolism
10.
Carcinogenesis ; 37(1): 72-80, 2016 Jan.
Article in English | MEDLINE | ID: mdl-26586792

ABSTRACT

Protein kinase C epsilon (PKCε), a Ca(2+)-independent phospholipid-dependent serine/threonine kinase, is among the six PKC isoforms (α, δ, ε, η, µ, ζ) expressed in both mouse and human skin. Epidermal PKCε level dictates the susceptibility of PKCε transgenic (TG) mice to the development of cutaneous squamous cell carcinomas (SCC) elicited either by repeated exposure to ultraviolet radiation (UVR) or by using the DMBA initiation-TPA (12-O-tetradecanoylphorbol-13-acetate) tumor promotion protocol (Wheeler,D.L. et al. (2004) Protein kinase C epsilon is an endogenous photosensitizer that enhances ultraviolet radiation-induced cutaneous damage and development of squamous cell carcinomas. Cancer Res., 64, 7756-7765). Histologically, SCC in TG mice, like human SCC, is poorly differentiated and metastatic. Our earlier studies to elucidate mechanisms of PKCε-mediated development of SCC, using either DMBA-TPA or UVR, indicated elevated release of cytokine TNFα. To determine whether TNFα is essential for the development of SCC in TG mice, we generated PKCε transgenic mice/TNFα-knockout (TG/TNFαKO) by crossbreeding TNFαKO with TG mice. We now present that deletion of TNFα in TG mice inhibited the development of SCC either by repeated UVR exposures or by the DMBA-TPA protocol. TG mice deficient in TNFα elicited both increase in SCC latency and decrease in SCC incidence. Inhibition of UVR-induced SCC development in TG/TNFαKO was accompanied by inhibition of (i) the expression levels of TNFα receptors TNFRI and TNFRII and cell proliferation marker ornithine decarboxylase and metastatic markers MMP7 and MMP9, (ii) the activation of transcription factors Stat3 and NF-kB and (iii) proliferation of hair follicle stem cells and epidermal hyperplasia. The results presented here provide the first genetic evidence that TNFα is linked to PKCε-mediated sensitivity to DMBA-TPA or UVR-induced development of cutaneous SCC.


Subject(s)
Carcinoma, Squamous Cell/prevention & control , Protein Kinase C-epsilon/genetics , Skin Neoplasms/prevention & control , Tumor Necrosis Factor-alpha/deficiency , Tumor Necrosis Factor-alpha/genetics , 9,10-Dimethyl-1,2-benzanthracene , Animals , Carcinogenesis/chemically induced , Carcinogenesis/genetics , Carcinogenesis/radiation effects , Carcinogens , Carcinoma, Squamous Cell/etiology , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/metabolism , Cell Survival/drug effects , Cell Survival/genetics , Cell Survival/radiation effects , Female , Male , Mice , Mice, Knockout , Mice, Transgenic , Protein Kinase C-epsilon/biosynthesis , Skin Neoplasms/etiology , Skin Neoplasms/genetics , Skin Neoplasms/metabolism , Tetradecanoylphorbol Acetate , Ultraviolet Rays
11.
Cancer Prev Res (Phila) ; 8(5): 375-86, 2015 May.
Article in English | MEDLINE | ID: mdl-25627799

ABSTRACT

Prostate cancer continues to remain the most common cancer and the second leading cause of cancer-related deaths in American males. The Pten deletions and/or mutations are frequently observed in both primary prostate cancers and metastatic prostate tissue samples. Pten deletion in prostate epithelium in mice results in prostatic intraepithelial neoplasia (PIN), followed by progression to invasive adenocarcinoma. The Pten conditional knockout mice [(Pten-loxp/loxp:PB-Cre4(+)) (Pten-KO)] provide a unique preclinical model to evaluate agents for efficacy for both the prevention and treatment of prostate cancer. We present here for the first time that dietary plumbagin, a medicinal plant-derived naphthoquinone (200 or 500 ppm) inhibits tumor development in intact as well as castrated Pten-KO mice. Plumbagin has shown no signs of toxicity at either of these doses. Plumbagin treatment resulted in a decrease expression of PKCε, AKT, Stat3, and COX2 compared with the control mice. Plumbagin treatment also inhibited the expression of vimentin and slug, the markers of epithelial-to-mesenchymal transition (EMT) in prostate tumors. In summary, the results indicate that dietary plumbagin inhibits growth of both primary and castration-resistant prostate cancer (CRPC) in Pten-KO mice, possibly via inhibition of PKCε, Stat3, AKT, and EMT markers (vimentin and slug), which are linked to the induction and progression of prostate cancer.


Subject(s)
Adenocarcinoma/prevention & control , Antineoplastic Agents, Phytogenic/pharmacology , Carcinogenesis/drug effects , Epithelial-Mesenchymal Transition/drug effects , Naphthoquinones/pharmacology , Prostatic Neoplasms/prevention & control , Adenocarcinoma/pathology , Animals , Antineoplastic Agents, Phytogenic/therapeutic use , Biomarkers/metabolism , Carcinogenesis/genetics , Carcinogenesis/metabolism , Epithelial-Mesenchymal Transition/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , Naphthoquinones/therapeutic use , Orchiectomy , PTEN Phosphohydrolase/genetics , Prostatic Intraepithelial Neoplasia/drug therapy , Prostatic Intraepithelial Neoplasia/pathology , Prostatic Neoplasms/pathology , Protein Kinase C-epsilon/metabolism , STAT3 Transcription Factor/metabolism , Signal Transduction/drug effects , Signal Transduction/genetics
12.
J Invest Dermatol ; 135(4): 1098-1107, 2015 Apr.
Article in English | MEDLINE | ID: mdl-25337691

ABSTRACT

We present here that heat-shock protein 90 (Hsp90) inhibitor 17-(allylamino)-17-demethoxygeldanamycin (17AAG), when topically applied to mouse skin, inhibits UVR-induced development of cutaneous squamous cell carcinoma (SCC). In these experiments, DMSO:acetone (1:40 v/v) solution of 17AAG (500 nmol) was applied topically to mouse skin in conjunction with each UVR exposure (1.8 kJ m(-2)). The UVR source was Kodacel-filtered FS-40 sun lamps (approximately 60% UVB and 40% UVA). In independent experiments with three separate mouse lines (SKH-1 hairless mice, wild-type FVB, and protein kinase C epsilon (PKCɛ)-overexpressing transgenic FVB mice), 17AAG treatment increased the latency and decreased both the incidence and multiplicity of UVR-induced SCC. Topical 17AAG alone or in conjunction with UVR treatments elicited neither skin nor systemic toxicity. 17AAG-caused inhibition of SCC induction was accompanied by a decrease in UVR-induced (1) hyperplasia, (2) Hsp90ß-PKCɛ interaction, and (3) expression levels of Hsp90ß, Stat3, pStat3Ser727, pStat3Tyr705, pAktSer473, and matrix metalloproteinase (MMP). The results presented here indicate that topical Hsp90 inhibitor 17AAG is effective in prevention of UVR-induced epidermal hyperplasia and SCC. One may conclude from the preclinical data presented here that topical 17AAG may be useful for prevention of UVR-induced inflammation and cutaneous SCC either developed in UVR-exposed or organ transplant population.


Subject(s)
Benzoquinones/pharmacology , HSP90 Heat-Shock Proteins/metabolism , Lactams, Macrocyclic/pharmacology , Melanoma/metabolism , Neoplasms, Radiation-Induced/metabolism , Skin Neoplasms/metabolism , Acetone/chemistry , Animals , Dimethyl Sulfoxide/chemistry , Epidermis/metabolism , HSP90 Heat-Shock Proteins/antagonists & inhibitors , Humans , Inflammation , Matrix Metalloproteinases/metabolism , Melanoma/prevention & control , Mice , Neoplasms, Radiation-Induced/prevention & control , Protein Kinase C-epsilon/metabolism , Skin/drug effects , Skin Neoplasms/drug therapy , Ultraviolet Rays , Melanoma, Cutaneous Malignant
13.
Antioxid Redox Signal ; 21(5): 682-99, 2014 Aug 10.
Article in English | MEDLINE | ID: mdl-24295217

ABSTRACT

AIMS: Pancreatic cancer (PC) is the most aggressive malignant disease, ranking as the fourth most leading cause of cancer-related death among men and women in the United States. In this study, we provide evidence of chemotherapeutic effects of α-mangostin, a dietary antioxidant isolated from the pericarp of Garcinia mangostana L. against human PC. RESULTS: The chemotherapeutic effect of α-mangostin was determined using four human PC cells (PL-45, PANC1, BxPC3, and ASPC1). α-Mangostin resulted in a significant inhibition of PC cells viability without having any effects on normal human pancreatic duct epithelial cells. α-Mangostin showed a dose-dependent increase of apoptosis in PC cells. Also, α-mangostin inhibited the expression levels of pNF-κB/p65Ser552, pStat3Ser727, and pStat3Tyr705. α-Mangostin inhibited DNA binding activity of nuclear factor kappa B (NF-κB) and signal transducer and activator 3 (Stat3). α-Mangostin inhibited the expression levels of matrix metallopeptidase 9 (MMP9), cyclin D1, and gp130; however, increased expression of tissue inhibitor of metalloproteinase 1 (TIMP1) was observed in PC cells. In addition, i.p. administration of α-mangostin (6 mg/kg body weight, 5 days a week) resulted in a significant inhibition of both primary (PL-45) and secondary (ASPC1) human PC cell-derived orthotopic and ectopic xenograft tumors in athymic nude mice. No sign of toxicity was observed in any of the mice administered with α-mangostin. α-Mangostin treatment inhibited the biomarkers of cell proliferation (Ki-67 and proliferating cell nuclear antigen [PCNA]) in the xenograft tumor tissues. INNOVATION: We present, for the first time, that dietary antioxidant α-mangostin inhibits the growth of PC cells in vitro and in vivo. CONCLUSION: These results suggest the potential therapeutic efficacy of α-mangostin against human PC.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , Antioxidants/pharmacology , Disease Models, Animal , Garcinia mangostana/chemistry , Pancreatic Neoplasms/drug therapy , Xanthones/pharmacology , Animals , Antineoplastic Agents, Phytogenic/administration & dosage , Antineoplastic Agents, Phytogenic/isolation & purification , Antioxidants/administration & dosage , Antioxidants/isolation & purification , Cell Line , Cell Proliferation/drug effects , Dose-Response Relationship, Drug , Humans , Injections, Intraperitoneal , Mice , Mice, Nude , Pancreatic Neoplasms/pathology , Structure-Activity Relationship , Xanthones/administration & dosage , Xanthones/isolation & purification , Xenograft Model Antitumor Assays
15.
J Skin Cancer ; 2013: 452425, 2013.
Article in English | MEDLINE | ID: mdl-23738074

ABSTRACT

To find clues about the mechanism by which kinase C epsilon (PKC ε ) may impart susceptibility to ultraviolet radiation (UVR)-induced development of cutaneous squamous cell carcinomas (SCC), we compared PKC ε transgenic (TG) mice and their wild-type (WT) littermates for (1) the effects of UVR exposures on percent of putative hair follicle stem cells (HSCs) and (2) HSCs proliferation. The percent of double HSCs (CD34+ and α 6-integrin or CD34+/CD49f+) in the isolated keratinocytes were determined by flow cytometric analysis. Both single and chronic UVR treatments (1.8 kJ/m(2)) resulted in an increase in the frequency of double positive HSCs in PKC ε TG mice as compared to their WT littermates. To determine the rate of proliferation of bulge region stem cells, a 5-bromo-2'-deoxyuridine labeling (BrdU) experiment was performed. In the WT mice, the percent of double positive HSCs retaining BrdU label was 28.4 ± 0.6% compared to 4.0 ± 0.06% for the TG mice, an approximately 7-fold decrease. A comparison of gene expression profiles of FACS sorted double positive HSCs showed increased expression of Pes1, Rad21, Tfdp1 and Cks1b genes in TG mice compared to WT mice. Also, PKC ε over expression in mice increased the clonogenicity of isolated keratinocytes, a property commonly ascribed to stem cells.

16.
Appl Biochem Biotechnol ; 170(4): 743-55, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23609909

ABSTRACT

Piriformospora indica is an axenically cultivable phytopromotional endosymbiont that mimics capabilities of arbuscular mycorrhizal fungi. This is a basidiomycete of the Sebacinaceae family, which promotes growth, development, and seed production in a variety of plant species. We report that the cell wall extract (CWE) from P. indica induces tuberization in vitro and promotes tuber growth and yield in potato. The CWE altered the calcium signaling pathway that regulates tuberization process. An increase in tuber number and size was correlated with increased transcript expression of the two Ca(2+)-dependant proteins (CaM1 and St-CDPK1) and the lipoxygenase (LOX) mRNA, which are known to play distinct roles in potato tuberization. External supplementation of Ca(2+) ions induced a similar set of tuberization pathway genes, indicating presence of an active Ca(2+) in the CWE of P. indica. Since potato tuberization is directly influenced by the presence of microflora in nature, the present study provides an insight into the novel mechanism of potato tuberization in relation to plant-microbe association. Ours is the first report on an in vitro tuber-inducing beneficial fungus.


Subject(s)
Basidiomycota/chemistry , Calcium Signaling , Cell Wall/chemistry , Plant Tubers/drug effects , Solanum tuberosum/drug effects , Biological Factors/pharmacology , Calcium/pharmacology , Culture Media/chemistry , Enzyme Activation , Gene Expression Regulation, Plant , Genes, Plant , Host-Pathogen Interactions , Lipoxygenase/genetics , Lipoxygenase/metabolism , Plant Proteins/genetics , Plant Tubers/enzymology , Plant Tubers/genetics , Plant Tubers/growth & development , RNA, Messenger/genetics , RNA, Messenger/metabolism , Solanum tuberosum/enzymology , Solanum tuberosum/genetics , Solanum tuberosum/growth & development
17.
Mol Oncol ; 7(3): 428-39, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23273564

ABSTRACT

We present here first time that Plumbagin (PL), a medicinal plant-derived 1,4-naphthoquinone, inhibits the growth and metastasis of human prostate cancer (PCa) cells in an orthotopic xenograft mouse model. In this study, human PCa PC-3M-luciferase cells (2 × 10(6)) were injected into the prostate of athymic nude mice. Three days post cell implantation, mice were treated with PL (2 mg/kg body wt. i.p. five days in a week) for 8 weeks. Growth and metastasis of PC-3M-luciferase cells was examined weekly by bioluminescence imaging of live mice. PL-treatment significantly (p = 0.0008) inhibited the growth of orthotopic xenograft tumors. Results demonstrated a significant inhibition of metastasis into liver (p = 0.037), but inhibition of metastasis into the lungs (p = 0.60) and lymph nodes (p = 0.27) was not observed to be significant. These results were further confirmed by histopathology of these organs. Results of histopathology demonstrated a significant inhibition of metastasis into lymph nodes (p = 0.034) and lungs (p = 0.028), and a trend to significance in liver (p = 0.075). None of the mice in the PL-treatment group showed PCa metastasis into the liver, but these mice had small metastasis foci into the lymph nodes and lungs. However, control mice had large metastatic foci into the lymph nodes, lungs, and liver. PL-caused inhibition of the growth and metastasis of PC-3M cells accompanies inhibition of the expression of: 1) PKCε, pStat3Tyr705, and pStat3Ser727, 2) Stat3 downstream target genes (survivin and Bcl(xL)), 3) proliferative markers Ki-67 and PCNA, 4) metastatic marker MMP9, MMP2, and uPA, and 5) angiogenesis markers CD31 and VEGF. Taken together, these results suggest that PL inhibits tumor growth and metastasis of human PCa PC3-M-luciferase cells, which could be used as a therapeutic agent for the prevention and treatment of human PCa.


Subject(s)
Antineoplastic Agents, Phytogenic/therapeutic use , Naphthoquinones/therapeutic use , Neoplasm Metastasis/prevention & control , Plumbaginaceae/chemistry , Prostate/pathology , Prostatic Neoplasms/drug therapy , Prostatic Neoplasms/pathology , Animals , Antineoplastic Agents, Phytogenic/chemistry , Antineoplastic Agents, Phytogenic/isolation & purification , Cell Line, Tumor , Gene Expression Regulation, Neoplastic/drug effects , Humans , Male , Matrix Metalloproteinase 2/genetics , Matrix Metalloproteinase 9/genetics , Mice , Mice, Nude , Naphthoquinones/chemistry , Naphthoquinones/isolation & purification , Neoplasm Metastasis/pathology , Nitric Oxide Synthase Type II/genetics , Prostate/drug effects , Prostate/metabolism , Prostatic Neoplasms/genetics , Protein Kinase C/genetics , STAT3 Transcription Factor/genetics
18.
Cancer Prev Res (Phila) ; 5(12): 1368-74, 2012 Dec.
Article in English | MEDLINE | ID: mdl-23060038

ABSTRACT

Decreasing the incidence of nonmelanoma skin cancer (NMSC) is of great importance in regards to future healthcare services. Given the previously reported preventive effects of α-difluoromethylornithine (DFMO) in skin and colon cancer trials, we determined appropriate cause to update the clinical data on the subjects from the recently reported randomized, double-blind, placebo-controlled phase III skin cancer prevention study of DFMO. Our intention was to retrospectively assess the further incidence of skin cancer, other malignancies, and adverse events of patients accrued to our phase III skin cancer prevention study of DFMO. Clinical records of 209 University of Wisconsin (UW) Health subjects were reviewed, and 2,092.7 person years of on study (884.3 person years) and poststudy (1,208.4 person years) follow-up for these patients were assessed for new NMSC events and recurrence rates from the on study period, the poststudy period, and the two study periods combined. No evidence of increased significant diagnoses or serious adverse events was observed in the DFMO participants. The initially observed, marginally significant reduction (P = 0.069) in NMSC rates for DFMO subjects relative to placebo continued without evidence of rebound. Event rates after discontinuation from study for total NMSCs (DFMO 0.236 NMSC/person/year, placebo 0.297, P = 0.48) or the subtypes of basal cell carcinomas (BCC; DFMO 0.179 BCC/person/year, placebo 0.190, P = 0.77) and squamous cell carcinomas (SCC; DFMO 0.057 SCC/person/year, placebo 0.107, P = 0.43) are listed. Follow-up data revealed a persistent but insignificant reduction in new NMSCs occurring in DFMO subjects without evidence of latent or cumulative toxicity relative to placebo subjects.


Subject(s)
Eflornithine/pharmacology , Skin Neoplasms/prevention & control , Adult , Aged , Carcinoma, Basal Cell/epidemiology , Carcinoma, Basal Cell/prevention & control , Carcinoma, Squamous Cell/epidemiology , Carcinoma, Squamous Cell/prevention & control , Double-Blind Method , Female , Follow-Up Studies , Humans , Incidence , Male , Middle Aged , Placebos , Skin Neoplasms/epidemiology , Wisconsin
19.
Carcinogenesis ; 33(12): 2586-92, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22976928

ABSTRACT

Plumbagin (PL), 5-hydroxy-2-methyl-1,4-naphthoquinone, is a quinoid constituent isolated from the roots of the medicinal plant Plumbago zeylanica L. (also known as chitrak). PL has also been found in Juglans regia (English Walnut), Juglans cinerea (whitenut) and Juglans nigra (blacknut). The roots of P. zeylanica have been used in Indian and Chinese systems of medicine for more than 2500 years for the treatment of various types of ailments. We were the first to report that PL inhibits the growth and invasion of hormone refractory prostate cancer (PCa) cells [Aziz,M.H. et al. (2008) Plumbagin, a medicinal plant-derived naphthoquinone, is a novel inhibitor of the growth and invasion of hormone-refractory prostate cancer. Cancer Res., 68, 9024-9032.]. Now, we present that PL inhibits in vivo PCa development in the transgenic adenocarcinoma of mouse prostate (TRAMP). PL treatment (2 mg/kg body weight i.p. in 0.2 ml phosphate-buffered saline, 5 days a week) to FVB-TRAMP resulted in a significant (P < 0.01) decrease in prostate tumor size and urogenital apparatus weights at 13 and 20 weeks. Histopathological analysis revealed that PL treatment inhibited progression of prostatic intraepithelial neoplasia (PIN) to poorly differentiated carcinoma (PDC). No animal exhibited diffuse tumor formation in PL-treated group at 13 weeks, whereas 75% of the vehicle-treated mice elicited diffuse PIN and large PDC at this stage. At 20 weeks, 25% of the PL-treated animals demonstrated diffuse PIN and 75% developed small PDC, whereas 100% of the vehicle-treated mice showed large PDC. PL treatment inhibited expression of protein kinase C epsilon (PKCε), signal transducers and activators of transcription 3 phosphorylation, proliferating cell nuclear antigen and neuroendocrine markers (synaptophysin and chromogranin-A) in excised prostate tumor tissues. Taken together, these results further suggest PL could be a novel chemopreventive agent against PCa.


Subject(s)
Adenocarcinoma/prevention & control , Anticarcinogenic Agents/therapeutic use , Chromogranin A/antagonists & inhibitors , Naphthoquinones/therapeutic use , Prostatic Neoplasms/prevention & control , Protein Kinase C-epsilon/antagonists & inhibitors , STAT3 Transcription Factor/antagonists & inhibitors , Synaptophysin/antagonists & inhibitors , Adenocarcinoma/pathology , Animals , Antigens, Polyomavirus Transforming/analysis , Disease Models, Animal , Male , Mice , Mice, Transgenic , Phosphorylation , Proliferating Cell Nuclear Antigen/analysis , Prostatic Neoplasms/pathology
20.
Int J Cancer ; 131(9): 2175-86, 2012 Nov 01.
Article in English | MEDLINE | ID: mdl-22322442

ABSTRACT

Pancreatic cancer (PC) is the most aggressive malignant disease, ranks as the fourth most leading cause of cancer-related death among men and women in the United States. We present here that plumbagin (PL), a quinoid constituent isolated from the roots of the medicinal plant Plumbago zeylanica L, inhibits the growth of PC cells both in vitro and in vivo model systems. PL treatment induces apoptosis and inhibits cell viability of PC cells (PANC1, BxPC3 and ASPC1). In addition, i.p. administration of PL (2 mg/kg body weight, 5 days a week) in severe combined immunodeficiency (SCID) mice beginning 3 days after ectopic implantation of PANC1 cells resulted in a significant (P < 0.01) inhibition of both tumor weight and volume. PL treatment inhibited (1) constitutive expression of epidermal growth factor receptor (EGFR), pStat3Tyr705 and pStat3Ser727, (2) DNA binding of Stat3 and (3) physical interaction of EGFR with Stat3, in both cultured PANC1 cells and their xenograft tumors. PL treatment also inhibited phosphorylation and DNA-binding activity of NF-κB in both cultured PC cells (PANC1 and ASPC1) and in PANC1 cells xenograft tumors. Downstream target genes (cyclin D1, MMP9 and Survivin) of Stat3 and NF-κB were similarly inhibited. These results suggest that PL may be used as a novel therapeutic agent against human PC. Published 2012 Wiley-Liss, Inc. This article is a US Government work, and, as such, is in the public domain in the United States of America.


Subject(s)
Antineoplastic Agents, Phytogenic/pharmacology , ErbB Receptors/metabolism , NF-kappa B/metabolism , Naphthoquinones/pharmacology , Pancreatic Neoplasms/drug therapy , Pancreatic Neoplasms/pathology , STAT3 Transcription Factor/metabolism , Animals , Apoptosis/drug effects , Cell Line, Tumor/drug effects , Cell Proliferation/drug effects , Cyclin D1/antagonists & inhibitors , Humans , Inhibitor of Apoptosis Proteins/antagonists & inhibitors , Male , Matrix Metalloproteinase Inhibitors , Mice , Mice, SCID , Phosphorylation , Plant Extracts/pharmacology , Signal Transduction/drug effects , Survivin
SELECTION OF CITATIONS
SEARCH DETAIL