Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 23
Filter
1.
Int J Nanomedicine ; 14: 7107-7121, 2019.
Article in English | MEDLINE | ID: mdl-31564868

ABSTRACT

BACKGROUND: Cervical cancer (CxCa) ranks as the fourth most prevalent women-related cancer worldwide. Therefore, there is a crucial need to develop newer treatment modalities. Ormeloxifene (ORM) is a non-steroidal, selective estrogen receptor modulator (SERM) that is used as an oral contraceptive in humans. Recent investigations suggest that ORM exhibits potent anti-cancer activity against various types of cancers. Nanoparticulates offer targeted delivery of anti-cancer drugs with minimal toxicity and promise newer approaches for cancer diagnosis and treatment. Therefore, the nanotherapy approach is superior compared to traditional chemotherapy, which is not site-specific and is often associated with various side effects. METHODS: Pursuing this novel nanotherapy approach, our lab has recently developed ORM-loaded poly [lactic-co-glycolic acid] (PLGA), an FDA-approved biodegradable polymer, nanoparticles to achieve targeted drug delivery and improved bioavailability. Our optimized PLGA-ORM nanoformulation showed improved internalization in both dose- and energy-dependent manners, through endocytosis-mediated pathways in both Caski and SiHa cell lines. Additionally, we employed MTS and colony forming assays to determine the short- and long-term effects of PLGA-ORM on these cells. RESULTS: Our results showed that this formulation demonstrated improved inhibition of cellular proliferation and clonogenic potential compared to free ORM. Furthermore, the PLGA-ORM nanoformulation exhibited superior anti-tumor activities in an orthotopic cervical cancer mouse model than free ORM. CONCLUSION: Collectively, our findings suggest that our novel nanoformulation has great potential for repurposing the drug and becoming a novel modality for CxCa management.


Subject(s)
Benzopyrans/therapeutic use , Nanoparticles/therapeutic use , Uterine Cervical Neoplasms/drug therapy , Animals , Benzopyrans/pharmacology , Carcinogenesis/drug effects , Carcinogenesis/metabolism , Carcinogenesis/pathology , Cell Line, Tumor , Cell Membrane/drug effects , Cell Membrane/metabolism , Cell Proliferation/drug effects , Cell Survival/drug effects , Clone Cells , Disease Models, Animal , Endocytosis/drug effects , Erythrocytes/metabolism , Female , Hemolysis/drug effects , Humans , Materials Testing , Membrane Potential, Mitochondrial/drug effects , Mice, Nude , Polylactic Acid-Polyglycolic Acid Copolymer/chemistry , Serum/chemistry , Uterine Cervical Neoplasms/pathology
2.
Sci Rep ; 9(1): 10917, 2019 07 29.
Article in English | MEDLINE | ID: mdl-31358785

ABSTRACT

Selective estrogen receptor modulator drug molecules of triphenylethylene family have gained considerable attention as anti-cancer agents. Despite recent advances in screening and development of HPV vaccines, cervical cancer remains one of the deadliest malignancies as advanced stage metastatic disease is mostly untreatable, thus warrants newer therapeutic strategies. Ormeloxifene (ORM) is a well-known SERM of triphenylethylene family that has been approved for human use, thus represents an ideal molecule for repurposing. In this study, we for the first time have demonstrated the anti-cancerous properties of ormeloxifene in cervical cancer. Ormeloxifene efficiently attenuated tumorigenic and metastatic properties of cervical cancer cells via arresting cell cycle at G1-S transition, inducing apoptosis, decreasing PI3K and Akt phosphorylation, mitochondrial membrane potential, and modulating G1-S transition related proteins (p21, cyclin E and Cdk2). Moreover, ORM repressed the expression of HPV E6/ E7 oncoproteins and restored the expression of their downstream target tumor suppressor proteins (p53, Rb and PTPN 13). As a result, ormeloxifene induces radio-sensitization in cervical cancer cells and caused potent tumor growth inhibition in orthotopic mouse model. Taken together, ormeloxifene represents an alternative therapeutic modality for cervical cancer which may have rapid clinical translation as it is already proven safe for human use.


Subject(s)
Antineoplastic Agents/therapeutic use , Benzopyrans/therapeutic use , Drug Repositioning , Uterine Cervical Neoplasms/drug therapy , Animals , Apoptosis/drug effects , Carcinogenesis/drug effects , Cell Line, Tumor , Female , G1 Phase Cell Cycle Checkpoints/drug effects , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Nude , Papillomavirus E7 Proteins/metabolism , Xenograft Model Antitumor Assays
3.
Cancer Res ; 77(14): 3766-3777, 2017 07 15.
Article in English | MEDLINE | ID: mdl-28490517

ABSTRACT

Medulloblastoma arising from the cerebellum is the most common pediatric brain malignancy, with leptomeningeal metastases often present at diagnosis and recurrence associated with poor clinical outcome. In this study, we used mouse medulloblastoma models to explore the relationship of tumor pathophysiology and dysregulated expression of the NOTCH pathway transcription factor ATOH1, which is present in aggressive medulloblastoma subtypes driven by aberrant Sonic Hedgehog/Patched (SHH/PTCH) signaling. In experiments with conditional ATOH1 mouse mutants crossed to Ptch1+/- mice, which develop SHH-driven medulloblastoma, animals with Atoh1 transgene expression developed highly penetrant medulloblastoma at a young age with extensive leptomeningeal disease and metastasis to the spinal cord and brain, resembling xenografts of human SHH medulloblastoma. Metastatic tumors retained abnormal SHH signaling like tumor xenografts. Conversely, ATOH1 expression was detected consistently in recurrent and metastatic SHH medulloblastoma. Chromatin immunoprecipitation sequencing and gene expression profiling identified candidate ATOH1 targets in tumor cells involved in development and tumorigenesis. Among these targets specific to metastatic tumors, there was an enrichment in those implicated in extracellular matrix remodeling activity, cytoskeletal network and interaction with microenvironment, indicating a shift in transcriptomic and epigenomic landscapes during metastasis. Treatment with bone morphogenetic protein or SHH pathway inhibitors decreased tumor cell proliferation and suppressed metastatic tumor growth, respectively. Our work reveals a dynamic ATOH1-driven molecular cascade underlying medulloblastoma metastasis that offers possible therapeutic opportunities. Cancer Res; 77(14); 3766-77. ©2017 AACR.


Subject(s)
Basic Helix-Loop-Helix Transcription Factors/biosynthesis , Cerebellar Neoplasms/metabolism , Medulloblastoma/metabolism , Medulloblastoma/pathology , Animals , Basic Helix-Loop-Helix Transcription Factors/genetics , Basic Helix-Loop-Helix Transcription Factors/metabolism , Cell Proliferation , Cerebellar Neoplasms/genetics , Cerebellar Neoplasms/pathology , Hedgehog Proteins , Heterografts , Humans , Medulloblastoma/genetics , Mice , Mice, Transgenic , Neoplasm Metastasis , Signal Transduction
4.
Sci Rep ; 6: 20051, 2016 Feb 03.
Article in English | MEDLINE | ID: mdl-26837852

ABSTRACT

Cervical cancer is one of the most common cancers among women worldwide. Current standards of care for cervical cancer includes surgery, radiation, and chemotherapy. Conventional chemotherapy fails to elicit therapeutic responses and causes severe systemic toxicity. Thus, developing a natural product based, safe treatment modality would be a highly viable option. Curcumin (CUR) is a well-known natural compound, which exhibits excellent anti-cancer potential by regulating many proliferative, oncogenic, and chemo-resistance associated genes/proteins. However, due to rapid degradation and poor bioavailability, its translational and clinical use has been limited. To improve these clinically relevant parameters, we report a poly(lactic-co-glycolic acid) based curcumin nanoparticle formulation (Nano-CUR). This study demonstrates that in comparison to free CUR, Nano-CUR effectively inhibits cell growth, induces apoptosis, and arrests the cell cycle in cervical cancer cell lines. Nano-CUR treatment modulated entities such as miRNAs, transcription factors, and proteins associated with carcinogenesis. Moreover, Nano-CUR effectively reduced the tumor burden in a pre-clinical orthotopic mouse model of cervical cancer by decreasing oncogenic miRNA-21, suppressing nuclear ß-catenin, and abrogating expression of E6/E7 HPV oncoproteins including smoking compound benzo[a]pyrene (BaP) induced E6/E7 and IL-6 expression. These superior pre-clinical data suggest that Nano-CUR may be an effective therapeutic modality for cervical cancer.


Subject(s)
Antineoplastic Agents/administration & dosage , Curcumin/administration & dosage , Lactic Acid/chemistry , Polyglycolic Acid/chemistry , Uterine Cervical Neoplasms/drug therapy , Animals , Antineoplastic Agents/chemistry , Antineoplastic Agents/pharmacology , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Curcumin/chemistry , Curcumin/pharmacology , Female , Gene Expression Regulation, Neoplastic/drug effects , Humans , Lactic Acid/administration & dosage , Mice , MicroRNAs/genetics , Nanoparticles/administration & dosage , Nanoparticles/chemistry , Polyglycolic Acid/administration & dosage , Polylactic Acid-Polyglycolic Acid Copolymer , Uterine Cervical Neoplasms/genetics , Xenograft Model Antitumor Assays
5.
Cancer Lett ; 356(2 Pt B): 606-12, 2015 Jan 28.
Article in English | MEDLINE | ID: mdl-25306892

ABSTRACT

Ovarian cancer continues to be a leading cause of cancer related deaths for women. Anticancer agents effective against chemo-resistant cells are greatly needed for ovarian cancer treatment. Repurposing drugs currently in human use is an attractive strategy for developing novel cancer treatments with expedited translation into clinical trials. Therefore, we examined whether ormeloxifene (ORM), a non-steroidal Selective Estrogen Receptor Modulator (SERM) currently used for contraception, is therapeutically effective at inhibiting ovarian cancer growth. We report that ORM treatment inhibits cell growth and induces apoptosis in ovarian cancer cell lines, including cell lines resistant to cisplatin. Furthermore, ORM treatment decreases Akt phosphorylation, increases p53 phosphorylation, and modulates the expression and localization patterns of p27, cyclin E, cyclin D1, and CDK2. In a pre-clinical xenograft mouse ORM treatment significantly reduces tumorigenesis and metastasis. These results indicate that ORM effectively inhibits the growth of cisplatin resistant ovarian cancer cells. ORM is currently in human use and has an established record of patient safety. Our encouraging in vitro and pre-clinical in vivo findings indicate that ORM is a promising candidate for the treatment of ovarian cancer.


Subject(s)
Apoptosis/drug effects , Benzopyrans/pharmacology , Cell Proliferation/drug effects , Ovarian Neoplasms/drug therapy , Ovarian Neoplasms/pathology , Animals , Blotting, Western , Cell Cycle/drug effects , Female , Flow Cytometry , Humans , Membrane Potential, Mitochondrial/drug effects , Mice , Mice, Nude , Ovarian Neoplasms/metabolism , Tumor Cells, Cultured , Tumor Stem Cell Assay , Xenograft Model Antitumor Assays
6.
Biomaterials ; 35(30): 8635-48, 2014 Oct.
Article in English | MEDLINE | ID: mdl-25028336

ABSTRACT

Prostate cancer is the most commonly diagnosed cancer disease in men in the Unites States and its management remains a challenge in everyday oncology practice. Thus, advanced therapeutic strategies are required to treat prostate cancer patients. Curcumin (CUR) is a promising anticancer agent for various cancer types. The objective of this study was to evaluate therapeutic potential of novel poly(lactic-co-glycolic acid)- CUR nanoparticles (PLGA-CUR NPs) for prostate cancer treatment. Our results indicate that PLGA-CUR NPs efficiently internalize in prostate cancer cells and release biologically active CUR in cytosolic compartment of cells for effective therapeutic activity. Cell proliferation (MTS), clonogenic, and Western blot analyses reveal that PLGA-CUR NPs can effectively inhibit proliferation and colony formation ability of prostate cancer cells than free CUR. PLGA-CUR NPs showed superior tumor regression compared to CUR in xenograft mice. Further investigations reveal that PLGA-CUR NPs inhibit nuclear ß-catenin and AR expression in cells and in tumor xenograft tissues. It also suppresses STAT3 and AKT phosphorylation and leads to apoptosis via inhibition of key anti-apoptotic proteins, Mcl-1, Bcl-xL and caused induction of PARP cleavage. Additionally, significant downregulation of oncogenic miR21 and up-regulation of miR-205 was observed with PLGA-CUR NPs treatment as determined by RT-PCR and in situ hybridization analyses. A superior anti-cancer potential was attained with PSMA antibody conjugated PLGA-CUR NPs in prostate cancer cells and a significant tumor targeting of (131)I labeled PSMA antibody was achieved with PLGA-CUR NPs in prostate cancer xenograft mice model. In conclusion, PLGA-CUR NPs can significantly accumulate and exhibit superior anticancer activity in prostate cancer.


Subject(s)
Antineoplastic Agents/therapeutic use , Curcumin/therapeutic use , Nanoparticles/chemistry , Prostatic Neoplasms/drug therapy , Animals , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Curcumin/pharmacology , Endocytosis/drug effects , Humans , Male , Mice, Nude , MicroRNAs/genetics , MicroRNAs/metabolism , Nanoparticles/ultrastructure , Prostate-Specific Antigen/metabolism , Prostatic Neoplasms/genetics , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Tumor Microenvironment/drug effects , Xenograft Model Antitumor Assays , beta Catenin/metabolism
7.
J Gastroenterol ; 49(10): 1378-91, 2014 Oct.
Article in English | MEDLINE | ID: mdl-24097071

ABSTRACT

BACKGROUND: MUC13 is overexpressed and aberrantly localized in colon cancer tissue; however, the specific functions and regulation of MUC13 expression are unknown. METHODS: Stable cell lines with either overexpressed or suppressed MUC13 levels were analyzed to determine cell growth, colony formation, cell migration, and cell invasion assays. The molecular mechanisms involved in MUC13 regulation were elucidated via chromatin immunoprecipitation (ChIP) and analysis of interleukin 6 (IL6) treatments. Colon cancer tissues were analyzed by immunohistochemistry (IHC) for the protein levels of MUC13 and P-STAT5 in colon cancer cells. RESULTS: Overexpression of MUC13 increased cell growth, colony formation, cell migration, and invasion. In concordance, MUC13 silencing decreased these tumorigenic features. Overexpression of MUC13 also modulated various cancer-associated proteins, including telomerase reverse transcriptase, sonic hedgehog, B cell lymphoma murine like site 1, and GATA like transcription factor 1. Additionally, MUC13-overexpressing cells showed increased HER2 and P-ERK expression. ChIP analysis revealed binding of STAT5 to the predicted MUC13 promoter. IL6 treatment of colon cancer cells increased the expression of MUC13 via activation of the JAK2/STAT5 signaling pathway. Suppression of JAK2 and STAT5 signaling by chemical inhibitors abolished IL6-induced MUC13 expression. IHC analysis showed increased expression of both P-STAT5 and MUC13 in colon cancer as compared to adjacent normal tissue. CONCLUSIONS: The results of this study, for the first time, suggest functional roles of MUC13 in colon cancer progression and provide information regarding the regulation of MUC13 expression via JAK2/STAT5 which may reveal promising therapeutic approaches for colon cancer treatment.


Subject(s)
Colonic Neoplasms/metabolism , Mucins/physiology , Neoplasm Proteins/physiology , Cell Division/physiology , Cell Movement/physiology , Colonic Neoplasms/genetics , Colonic Neoplasms/pathology , Gene Expression Regulation, Neoplastic/drug effects , Gene Expression Regulation, Neoplastic/physiology , Gene Knockdown Techniques/methods , Humans , Interleukin-6/pharmacology , Mucins/biosynthesis , Mucins/genetics , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/biosynthesis , Neoplasm Proteins/genetics , STAT5 Transcription Factor/metabolism , STAT5 Transcription Factor/physiology , Signal Transduction/physiology , Tumor Cells, Cultured , Tumor Stem Cell Assay/methods , Up-Regulation/drug effects , Up-Regulation/physiology
8.
J Ovarian Res ; 5(1): 44, 2012 Dec 13.
Article in English | MEDLINE | ID: mdl-23237306

ABSTRACT

Ovarian cancer is the fifth most common cancer among women and causes more deaths than any other type of female reproductive cancer. Currently, treatment of ovarian cancer is based on the combination of surgery and chemotherapy. While recurrent ovarian cancer responds to additional chemotherapy treatments, the progression-free interval becomes shorter after each cycle, as chemo-resistance increases until the disease becomes incurable. There is, therefore, a strong need for prognostic and predictive markers to help optimize and personalize treatment in order to improve the outcome of ovarian cancer. An increasing number of studies indicate an essential role for microRNAs in ovarian cancer progression and chemo-resistance. MicroRNAs (miRNAs) are small endogenous non-coding RNAs (~22bp) which are frequently dysregulated in cancer. Typically, miRNAs are involved in crucial biological processes, including development, differentiation, apoptosis and proliferation. Two families of miRNAs, miR-200 and let-7, are frequently dysregulated in ovarian cancer and have been associated with poor prognosis. Both have been implicated in the regulation of epithelial-to-mesenchymal transition, a cellular transition associated with tumor aggressiveness, tumor invasion and chemo-resistance. Moreover, miRNAs also have possible implications for improving cancer diagnosis; for example miR-200 family, let-7 family, miR-21 and miR-214 may be useful in diagnostic tests to help detect ovarian cancer at an early stage. Additionally, the use of multiple target O-modified antagomirs (MTG-AMO) to inhibit oncogenic miRNAs and miRNA replacement therapy for tumor suppressor miRNAs are essential tools for miRNA based cancer therapeutics. In this review we describe the current status of the role miRNAs play in ovarian cancer and focus on the possibilities of microRNA-based therapies and the use of microRNAs as diagnostic tools.

9.
J Histochem Cytochem ; 60(11): 822-31, 2012 Nov.
Article in English | MEDLINE | ID: mdl-22914648

ABSTRACT

MUC13 is a newly identified transmembrane mucin. Although MUC13 is known to be overexpressed in ovarian and gastric cancers, limited information is available regarding the expression of MUC13 in metastatic colon cancer. Herein, we investigated the expression profile of MUC13 in colon cancer using a novel anti-MUC13 monoclonal antibody (MAb, clone ppz0020) by immunohistochemical (IHC) analysis. A cohort of colon cancer samples and tissue microarrays containing adjacent normal, non-metastatic colon cancer, metastatic colon cancer, and liver metastasis tissues was used in this study to investigate the expression pattern of MUC13. IHC analysis revealed significantly higher (p<0.001) MUC13 expression in non-metastatic colon cancer samples compared with faint or very low expression in adjacent normal tissues. Interestingly, metastatic colon cancer and liver metastasis tissue samples demonstrated significantly (p<0.05) higher cytoplasmic and nuclear MUC13 expression compared with non-metastatic colon cancer and adjacent normal colon samples. Moreover, cytoplasmic and nuclear MUC13 expression correlated with larger and poorly differentiated tumors. Four of six tested colon cancer cell lines also expressed MUC13 at RNA and protein levels. These studies demonstrate a significant increase in MUC13 expression in metastatic colon cancer and suggest a correlation between aberrant MUC13 localization (cytoplasmic and nuclear expression) and metastatic colon cancer.


Subject(s)
Colon/pathology , Colonic Neoplasms/pathology , Liver Neoplasms/secondary , Liver/pathology , Mucins/analysis , Cell Line, Tumor , Cell Nucleus/metabolism , Cell Nucleus/pathology , Cohort Studies , Colon/metabolism , Colonic Neoplasms/genetics , Cytoplasm/metabolism , Cytoplasm/pathology , Gene Expression Regulation, Neoplastic , Humans , Immunohistochemistry , Mucins/genetics , RNA/genetics , Tissue Array Analysis , Up-Regulation
10.
Curr Drug Metab ; 13(1): 120-8, 2012 Jan.
Article in English | MEDLINE | ID: mdl-21892919

ABSTRACT

Prostate cancer (PC) is the most frequently diagnosed disease in men in the United States. Curcumin (CUR), a natural diphenol, has shown potent anti-cancer efficacy in various types of cancers. However, suboptimal pharmacokinetics and poor bioavailability limit its effective use in cancer therapeutics. Several successful CUR nanoformulations have recently been reported which improve upon these features; however, there is no personalized safe nanoformulation for prostate cancer. This study contributes two important scientific aspects of prostate cancer therapeutics. The first objective was to investigate the comparative cellular uptake and cytotoxicity evaluation of ß-cyclodextrin (CD), hydroxypropyl methylcellulose (cellulose), poly(lactic-co-glycolic acid) (PLGA), magnetic nanoparticles (MNP), and dendrimer based CUR nanoformulations in prostate cancer cells. Curcumin loaded cellulose nanoparticles (cellulose-CUR) formulation exhibited the highest cellular uptake and caused maximum ultrastructural changes related to apoptosis (presence of vacuoles) in prostate cancer cells. Secondly, the anti-cancer potential of the cellulose-CUR formulation was evaluated in cell culture models using cell proliferation, colony formation and apoptosis (7-AAD staining) assays. In these assays, the cellulose-CUR formulation showed improved anti-cancer efficacy compared to free curcumin. Our study shows, for the first time, the feasibility of cellulose-CUR formulation and its potential use in prostate cancer therapy.


Subject(s)
Cellulose/administration & dosage , Curcumin/administration & dosage , Curcumin/chemistry , Nanoparticles/administration & dosage , Prostatic Neoplasms/drug therapy , Apoptosis/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Cellulose/adverse effects , Chemistry, Pharmaceutical/methods , Humans , Lactic Acid/administration & dosage , Lactic Acid/adverse effects , Magnetics/methods , Male , Polyglycolic Acid/administration & dosage , Polyglycolic Acid/adverse effects , Polylactic Acid-Polyglycolic Acid Copolymer , Prostatic Neoplasms/pathology , Tumor Stem Cell Assay/methods , beta-Cyclodextrins/administration & dosage , beta-Cyclodextrins/adverse effects
11.
Mol Cancer Ther ; 11(1): 24-33, 2012 Jan.
Article in English | MEDLINE | ID: mdl-22027689

ABSTRACT

The high death rate of pancreatic cancer is attributed to the lack of reliable methods for early detection and underlying molecular mechanisms of its aggressive pathogenesis. Although MUC13, a newly identified transmembrane mucin, is known to be aberrantly expressed in ovarian and gastro-intestinal cancers, its role in pancreatic cancer is unknown. Herein, we investigated the expression profile and functions of MUC13 in pancreatic cancer progression. The expression profile of MUC13 in pancreatic cancer was investigated using a recently generated monoclonal antibody (clone PPZ0020) and pancreatic tissue microarrays. The expression of MUC13 was significantly (P < 0.005) higher in cancer samples compared with normal/nonneoplastic pancreatic tissues. For functional analyses, full-length MUC13 was expressed in MUC13 null pancreatic cancer cell lines, MiaPaca and Panc1. MUC13 overexpression caused a significant (P < 0.05) increase in cell motility, invasion, proliferation, and anchorage-dependent or -independent clonogenicity while decreasing cell-cell and cell-substratum adhesion. Exogenous MUC13 expression significantly (P < 0.05) enhanced pancreatic tumor growth and reduced animal survival in a xenograft mouse model. These tumorigenic characteristics correlated with the upregulation/phosphorylation of HER2, p21-activated kinase 1 (PAK1), extracellular signal-regulated kinase (ERK), Akt, and metastasin (S100A4), and the suppression of p53. Conversely, suppression of MUC13 in HPAFII pancreatic cancer cells by short hairpin RNA resulted in suppression of tumorigenic characteristics, repression of HER2, PAK1, ERK, and S100A4, and upregulation of p53. MUC13 suppression also significantly (P < 0.05) reduced tumor growth and increased animal survival. These results imply a role of MUC13 in pancreatic cancer and suggest its potential use as a diagnostic and therapeutic target.


Subject(s)
Antibodies, Monoclonal/immunology , Mucins/metabolism , Pancreatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Cell Movement , Cell Proliferation , Cell Transformation, Neoplastic , Extracellular Signal-Regulated MAP Kinases/metabolism , Humans , Mice , Mice, Nude , Mucins/immunology , Neoplasm Invasiveness , Neoplasm Transplantation , RNA Interference , RNA, Small Interfering , Receptor, ErbB-2/metabolism , S100 Calcium-Binding Protein A4 , S100 Proteins/metabolism , Signal Transduction/physiology , Tumor Suppressor Protein p53/metabolism , p21-Activated Kinases/metabolism
12.
BMC Infect Dis ; 11: 252, 2011 Sep 24.
Article in English | MEDLINE | ID: mdl-21943050

ABSTRACT

BACKGROUND: High-risk strains of human papillomavirus (HPV) cause cervical cancer. American Indian (AI) women in the Northern Plains of the U.S. have significantly higher incidence and mortality rates for cervical cancer than White women in the same geographical area. We compared HPV prevalence, patterns of HPV types, and infection with multiple HPV types in AI and White women living in South Dakota, U.S. METHODS: We analyzed the HPV status of cervical samples collected in 2006-2008 from women aged 18-65 years who attended two rural AI reservation clinics (n = 235) or an urban clinic in the same area serving mostly White women (n = 246). Data collection occurred before HPV vaccination was available to study participants. HPV DNA was amplified by using the L1 consensus primer system and an HPV Linear Array detection assay to identify HPV types. We used chi-square tests to compare HPV variables, with percentages standardized by age and lifetime number of sexual partners. RESULTS: Compared to White women, AI women were younger (p = 0.01) and reported more sexual partners (p < 0.001). A lower percentage of AI women tested negative for HPV infection compared to Whites (58% [95% CI = 51-65] vs. 77% [95% CI = 71-82]; p < 0.001), and a higher percentage of AI women were infected by oncogenic types (30% [95% CI = 25-36] vs. 16% [95% CI = 11-21]; p = 0.001). Infections among AI women showed a wider variety and very different pattern of HPV types, including a higher prevalence of mixed HPV infections (19% [95% CI = 26-38] vs. 7% [95% CI = 4-11]; p = 0.001). AI women had a higher percentage of HPV infections that were not preventable by HPV vaccination (32% [95% CI = 26-38] vs. 15% [95% CI = 11-21]; p < 0.001). CONCLUSIONS: A higher HPV burden and a different HPV genotyping profile may contribute to the high rate of cervical cancer among AI women.


Subject(s)
Papillomaviridae/classification , Papillomaviridae/isolation & purification , Papillomavirus Infections/epidemiology , Papillomavirus Infections/virology , Adolescent , Adult , Aged , Female , Genotype , Humans , Indians, North American , Middle Aged , Papillomavirus Infections/genetics , Prevalence , Rural Population , South Dakota/epidemiology , Urban Population , White People , Young Adult
13.
Mol Cancer Res ; 9(5): 531-537, 2011 May.
Article in English | MEDLINE | ID: mdl-21450906

ABSTRACT

Mucin 13 (MUC13) is a high-molecular-weight transmembrane glycoprotein that is frequently and aberrantly expressed in a variety of epithelial carcinomas, including gastric, colorectal, and ovarian cancers. On the basis of the high expression of MUC13 in cancer cells as well as recent laboratory findings suggesting a malignant phenotype of MUC13-transfected cell lines, the oncogenic potential of MUC13 has emerged. The various functional domains of MUC13 may confer oncogenic potential to MUC13. For example, the bulky extracellular domain with extensive modification with glycan chains may prevent cell-cell and cell-extracellular matrix binding whereas the cytoplasmic tail containing serine and tyrosine residues for potential phosphorylation may participate in cell signaling. MUC13 exhibits the characteristics suitable as an early marker for cancer screening and presents a promising target for antibody-guided targeted therapy.


Subject(s)
Colorectal Neoplasms/metabolism , Mucins/chemistry , Mucins/metabolism , Ovarian Neoplasms/metabolism , Precancerous Conditions/metabolism , Stomach Neoplasms/metabolism , Amino Acid Sequence , Female , Glycosylation , Humans , Intracellular Signaling Peptides and Proteins/chemistry , Membrane Glycoproteins/biosynthesis , Membrane Glycoproteins/chemistry , Membrane Glycoproteins/metabolism , Molecular Sequence Data , Mucins/biosynthesis , Oncogene Proteins/chemistry , Oncogene Proteins/metabolism , Phosphorylation , Receptors, Cell Surface/chemistry , Receptors, Cell Surface/metabolism
14.
Anticancer Res ; 31(3): 897-906, 2011 Mar.
Article in English | MEDLINE | ID: mdl-21498711

ABSTRACT

AIM: The aim of this study was to prepare biotinylated PAMAM dendrimers loaded with cisplatin and to evaluate the cytotoxicity in ovarian cancer cell lines. MATERIALS AND METHODS: Biotinylated and unconjugated dendrimer-cisplatin complexes were investigated for encapsulation efficiency, in vitro cytotoxic activity and cellular accumulation of cisplatin in OVCAR-3, SKOV-3, A2780 (wild-type) and CP70 (A2780/CP70, cisplatin-resistant) cells. RESULTS: Encapsulation efficiency of cisplatin ranged from 5.33% to 21.10%. In vitro cytotoxic activity revealed that IC(50) values of dendrimer-cisplatin complexes were significantly lower than that of free cisplatin in OVCAR-3, SKOV-3 and CP70 cell lines. Cellular uptake data showed highest accumulation of platinum by PAMAMG(4) NH(2) dendrimer complexes of cisplatin in A2780 (19.41±0.85 µg/ml) and CP70 (25.25±1.25 µg/ml) cell lines in comparison with cisplatin uptake of only 1.77±0.351 µg/ml in A2780 and 2.31±0.421 µg/ml in CP70 cells. CONCLUSION: In conclusion, biotinylated PAMAM dendrimers may be utilized as potential targeting agents for cisplatin delivery to ovarian cancer.


Subject(s)
Cisplatin/pharmacology , Dendrimers/pharmacology , Drug Delivery Systems , Intracellular Space/metabolism , Ovarian Neoplasms/metabolism , Biotinylation/drug effects , Cell Death/drug effects , Cell Line, Tumor , Cell Survival/drug effects , Female , Fluorescein-5-isothiocyanate/metabolism , Gene Expression Regulation, Neoplastic/drug effects , Hemolysis/drug effects , Humans , Inhibitory Concentration 50 , Intracellular Space/drug effects , Materials Testing , Ovarian Neoplasms/genetics , Ovarian Neoplasms/pathology , Reverse Transcriptase Polymerase Chain Reaction , Symporters/genetics , Symporters/metabolism
15.
Gynecol Oncol ; 121(3): 532-6, 2011 Jun 01.
Article in English | MEDLINE | ID: mdl-21414655

ABSTRACT

OBJECTIVE: American Indian (AI) women living in the Northern Plains have high incidence and mortality rates for cervical cancer. We assessed risk factors for human papillomavirus (HPV) infection among AI and White women. METHODS: We tested cervical samples for HPV infection obtained from women ages 18-65 years attending 2 rural AI reservation clinics in South Dakota (n=235) and an urban clinic serving predominantly White women (n=246). Patients self-reported information on HPV risk factors. We used percentages and chi-square tests to compare risk factors, and logistic regression with HPV status as the outcome to quantify the association between HPV and risk factors. RESULTS: AI women had more risk factors than White women, including younger age, less education, less vegetable consumption, more sexual partners, younger age at first sexual experience and first pregnancy, and more pregnancies (p values≤0.003). AI women more often endorsed recreational drug use, history of sexually transmitted diseases, and current smoking; White women reported more alcohol consumption (p values<0.001). In multivariate analysis, younger age and current smoking were associated with higher odds of HPV infection in AI women, whereas a higher number of sexual partners was associated with higher odds of HPV infection in White women. CONCLUSIONS: AI women have a high burden of risk factors for HPV disease, and associations with HPV infection appear to differ by community. Knowledge of specific risk factors in AI populations may provide targets for public health officials to decrease HPV infection and disease.


Subject(s)
Indians, North American/statistics & numerical data , Papillomavirus Infections/ethnology , White People/statistics & numerical data , Adolescent , Adult , Age Factors , Cervix Uteri/virology , Female , Humans , Middle Aged , Papillomavirus Infections/epidemiology , Pregnancy , Risk Factors , Rural Population , Sexual Behavior , South Dakota/epidemiology , Urban Population , Young Adult
16.
Mol Carcinog ; 50(1): 47-57, 2011 Jan.
Article in English | MEDLINE | ID: mdl-21061268

ABSTRACT

Curcumin has great potential as a chemopreventive and chemotherapeutic agent; however, its effects on human papillomavirus (HPV)-associated molecular events are inadequately explored. This study examined the effects of curcumin on HPV-associated pathways involved in developing cervical cancer. We demonstrate for the first time that curcumin treatment suppresses cervical cancer cell growth in a three-dimensional raft culture system. Curcumin also inhibits tumorigenic characteristics as shown by decreases in both clonogenic potential and cell motility. Additionally, our findings show that curcumin treatment inhibits the transcription of HPV16 E6/E7 as early as 6 h posttreatment and restores the expression of tumor suppressor proteins p53, retinoblastoma protein, and PTPN13. While smoking is a recognized risk factor for cervical cancer, the molecular effects of smoke carcinogens on the expression of HPV E6/E7 oncogenes are not well known. We show for the first time that exposure to benzo[a]pyrene (BaP), a tobacco carcinogen, increases the expression of HPV E7 oncoprotein suggesting a molecular link between smoking and cervical cancer. Importantly, curcumin decreases the BaP induced increase in the expression of HPV E7 oncoprotein. The results of this study clearly demonstrate that curcumin alters HPV-associated molecular pathways in cervical cancer cells. These novel findings imply that curcumin may be an effective chemopreventive and therapeutic agent for cervical cancer prevention and treatment.


Subject(s)
Benzo(a)pyrene/pharmacology , Curcumin/pharmacology , Oncogene Proteins, Viral/antagonists & inhibitors , Papillomavirus E7 Proteins/antagonists & inhibitors , Protein Tyrosine Phosphatase, Non-Receptor Type 13/metabolism , Repressor Proteins/antagonists & inhibitors , Retinoblastoma Protein/metabolism , Tumor Suppressor Protein p53/metabolism , Uterine Cervical Neoplasms/metabolism , Antineoplastic Agents/pharmacology , Apoptosis/drug effects , Blotting, Western , Cell Adhesion/drug effects , Cell Movement/drug effects , Cell Proliferation/drug effects , Female , Flow Cytometry , Humans , Oncogene Proteins, Viral/genetics , Oncogene Proteins, Viral/metabolism , Papillomavirus E7 Proteins/genetics , Papillomavirus E7 Proteins/metabolism , Protein Tyrosine Phosphatase, Non-Receptor Type 13/genetics , RNA, Messenger/genetics , Repressor Proteins/genetics , Repressor Proteins/metabolism , Retinoblastoma Protein/genetics , Reverse Transcriptase Polymerase Chain Reaction , Tumor Cells, Cultured , Tumor Stem Cell Assay , Tumor Suppressor Protein p53/genetics , Up-Regulation , Uterine Cervical Neoplasms/drug therapy , Uterine Cervical Neoplasms/genetics
17.
J Ovarian Res ; 3: 11, 2010 Apr 29.
Article in English | MEDLINE | ID: mdl-20429876

ABSTRACT

BACKGROUND: Chemo/radio-resistance is a major obstacle in treating advanced ovarian cancer. The efficacy of current treatments may be improved by increasing the sensitivity of cancer cells to chemo/radiation therapies. Curcumin is a naturally occurring compound with anti-cancer activity in multiple cancers; however, its chemo/radio-sensitizing potential is not well studied in ovarian cancer. Herein, we demonstrate the effectiveness of a curcumin pre-treatment strategy for chemo/radio-sensitizing cisplatin resistant ovarian cancer cells. To improve the efficacy and specificity of curcumin induced chemo/radio sensitization, we developed a curcumin nanoparticle formulation conjugated with a monoclonal antibody specific for cancer cells. METHODS: Cisplatin resistant A2780CP ovarian cancer cells were pre-treated with curcumin followed by exposure to cisplatin or radiation and the effect on cell growth was determined by MTS and colony formation assays. The effect of curcumin pre-treatment on the expression of apoptosis related proteins and beta-catenin was determined by Western blotting or Flow Cytometry. A luciferase reporter assay was used to determine the effect of curcumin on beta-catenin transcription activity. The poly(lactic acid-co-glycolic acid) (PLGA) nanoparticle formulation of curcumin (Nano-CUR) was developed by a modified nano-precipitation method and physico-chemical characterization was performed by transmission electron microscopy and dynamic light scattering methods. RESULTS: Curcumin pre-treatment considerably reduced the dose of cisplatin and radiation required to inhibit the growth of cisplatin resistant ovarian cancer cells. During the 6 hr pre-treatment, curcumin down regulated the expression of Bcl-XL and Mcl-1 pro-survival proteins. Curcumin pre-treatment followed by exposure to low doses of cisplatin increased apoptosis as indicated by annexin V staining and cleavage of caspase 9 and PARP. Additionally, curcumin pre-treatment lowered beta-catenin expression and transcriptional activity. Nano-CUR was successfully generated and physico-chemical characterization of Nano-CUR indicated an average particle size of ~70 nm, steady and prolonged release of curcumin, antibody conjugation capability and effective inhibition of ovarian cancer cell growth. CONCLUSION: Curcumin pre-treatment enhances chemo/radio-sensitization in A2780CP ovarian cancer cells through multiple molecular mechanisms. Therefore, curcumin pre-treatment may effectively improve ovarian cancer therapeutics. A targeted PLGA nanoparticle formulation of curcumin is feasible and may improve the in vivo therapeutic efficacy of curcumin.

18.
PLoS One ; 3(4): e2067, 2008 Apr 30.
Article in English | MEDLINE | ID: mdl-18446191

ABSTRACT

BACKGROUND: Candida albicans is a low level commensal organism in normal human populations with the continuous potential to expand and cause a spectrum of clinical conditions. METHODOLOGY/PRINCIPAL FINDINGS: Using ex vivo human organ cultures and populations of primary human cells, we have developed several related experimental systems to examine early-stage interactions between C. albicans and mucosal surfaces. Experiments have been conducted both with exogenously added C. albicans and with overtly normal human mucosal surfaces supporting pre-existing infections with natural isolates of Candida. Under different culture conditions, we have demonstrated the formation of C. albicans colonies on human target cells and filament formation, equivalent to tissue invasion. CONCLUSIONS/SIGNIFICANCE: These organ culture systems provide a valuable new resource to examine the molecular and cellular basis for Candida colonization of human mucosal surfaces.


Subject(s)
Candida albicans/growth & development , Colony Count, Microbial , Mucous Membrane/microbiology , Adolescent , Adult , Aged , Candida albicans/isolation & purification , Candidiasis/microbiology , Cell Proliferation , Cells, Cultured , Child, Preschool , Female , Humans , Imaging, Three-Dimensional , Male , Microscopy, Confocal , Mucous Membrane/cytology , Mucous Membrane/pathology , Palatine Tonsil/microbiology , Palatine Tonsil/pathology
19.
J Histochem Cytochem ; 55(8): 867-75, 2007 Aug.
Article in English | MEDLINE | ID: mdl-17478446

ABSTRACT

Single antigen-targeted intraperitoneal radioimmunotherapy for ovarian cancer has shown limited success. Due to the heterogeneous expression of tumor antigens on cancer cells, a multi-antigen targeting approach appears logical to augment the therapeutic efficacy of antibody-guided therapy. In the interest of developing this novel approach, ovarian cancer tissue microarray slides containing cancer and benign/non-neoplastic tissue samples (n=92) were processed for single-, double-, and triple-antigen labeling using antibodies for the tumor-associated antigens TAG-72, MUC1, and CA125. Among all ovarian cancer types, 72%, 61%, and 50% of the samples showed immunolabeling for TAG-72, MUC1, and CA125, respectively. Expression level of these antigens was significantly (p<0.005) higher in advanced stage carcinomas compared with early stage. Of the 48 epithelial ovarian cancer samples, individual anti-TAG-72, MUC1, and CA125 antibody probing showed labeling in 89.5%, 87.5%, and 73.0% of the cases, respectively. In the majority of the cancer samples (>70%), a heterogeneous labeling pattern was observed (only 30-40% of the cancer cells within the sample were labeled). However, upon combining the three antigens (triple-antigen labeling), 98% of the epithelial ovarian cancer samples were labeled and >95% of the cancer cells within each sample were labeled. Our data indicate that the heterogeneous expression of cancer antigens appears to be a major obstacle in antibody-guided therapy, and this can be overcome by multiple antigen targeting. Therapeutic efficacy of antibody-guided therapy for ovarian cancer treatment will be enhanced by the combined targeting of TAG-72, MUC1, and CA125.


Subject(s)
Antibodies , Antigens, Neoplasm/metabolism , Biomarkers, Tumor/metabolism , CA-125 Antigen/metabolism , Glycoproteins/metabolism , Mucins/metabolism , Ovarian Neoplasms/metabolism , Drug Carriers , Female , Humans , Mucin-1 , Neoplasms, Glandular and Epithelial/metabolism , Ovarian Neoplasms/pathology , Ovarian Neoplasms/radiotherapy , Ovary/metabolism , Tissue Array Analysis
20.
Virol J ; 3: 25, 2006 Apr 06.
Article in English | MEDLINE | ID: mdl-16600047

ABSTRACT

BACKGROUND: Primary HIV infection can develop from exposure to HIV in the oral cavity. In previous studies, we have documented rapid and extensive binding of HIV virions in seminal plasma to intact mucosal surfaces of the palatine tonsil and also found that virions readily penetrated beneath the tissue surfaces. As one approach to understand the molecular interactions that support HIV virion binding to human mucosal surfaces, we have examined the distribution of the primary HIV receptor CD4, the alternate HIV receptors heparan sulfate proteoglycan (HS) and galactosyl ceramide (GalCer) and the co-receptors CXCR4 and CCR5 in palatine tonsil. RESULTS: Only HS was widely expressed on the surface of stratified squamous epithelium. In contrast, HS, GalCer, CXCR4 and CCR5 were all expressed on the reticulated epithelium lining the tonsillar crypts. We have observed extensive variability, both across tissue sections from any tonsil and between tonsils, in the distribution of epithelial cells expressing either CXCR4 or CCR5 in the basal and suprabasal layers of stratified epithelium. The general expression patterns of CXCR4, CCR5 and HS were similar in palatine tonsil from children and adults (age range 3-20). We have also noted the presence of small clusters of lymphocytes, including CD4+ T cells within stratified epithelium and located precisely at the mucosal surfaces. CD4+ T cells in these locations would be immediately accessible to HIV virions. CONCLUSION: In total, the likelihood of oral HIV transmission will be determined by macro and micro tissue architecture, cell surface expression patterns of key molecules that may bind HIV and the specific properties of the infectious inoculum.


Subject(s)
HIV Infections/etiology , Mouth Diseases/etiology , Palatine Tonsil/virology , Receptors, HIV/analysis , Epithelial Cells/chemistry , Galactosylceramides/analysis , Heparan Sulfate Proteoglycans/analysis , Humans , Immunohistochemistry , Intercellular Adhesion Molecule-1/analysis , Interleukin-8/analysis , Lymphocyte Function-Associated Antigen-1/analysis , Palatine Tonsil/chemistry , Palatine Tonsil/immunology , Receptors, CCR5/analysis , Receptors, CXCR4/analysis , T-Lymphocyte Subsets/immunology
SELECTION OF CITATIONS
SEARCH DETAIL
...