Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 98
Filter
Add more filters

Country/Region as subject
Publication year range
1.
J Minim Access Surg ; 19(3): 427-432, 2023.
Article in English | MEDLINE | ID: mdl-37470632

ABSTRACT

Introduction: The aim of this study was to compare the peri-operative outcomes, especially intraoperative surgeon workload and early post-operative pain, following midline ventral hernia repair by laparoscopic enhanced-view totally extraperitoneal (eTEP) approach and laparoscopic intraperitoneal onlay mesh plus (IPOM plus) approach. Patients and Methods: This single-centre randomised control trial study was conducted from January 2020 to June 2022. A total of 60 adult patients undergoing elective ventral hernia surgery with small- and medium-sized midline defects were included. Intraoperative surgeon workload and early post-operative pain were systematically recorded and analysed for each procedure. Results: Out of 30 patients assigned to each group, 29 patients underwent eTEP mesh repair and 27 patients underwent successful IPOM plus repair. The intraoperative surgeon's workload, especially mental demand, physical demand, task complexity and degree of difficulty as reported and felt by the operating surgeon, was significantly higher in the eTEP mesh repair group compared to IPOM plus group (P < 0.001) with comparable operating room distractions (P = 0.039). The mean overall post-operative pain score on post-operative day 1 was slightly less in eTEP mesh repair (4.28 ± 1.12) group compared to IPOM plus group (4.93 ± 1.17), which was statistically insignificant (P = 0.042). The eTEP group had significantly longer operative time and length of hospital stay compared to the IPOM plus group. Conclusion: Our study revealed significantly longer operative time, higher surgical workload and increased length of hospital stay in the eTEP group with comparable early post-operative pain in both groups, thus making eTEP mesh repair a more difficult and challenging procedure.

2.
Adv Exp Med Biol ; 890: 57-74, 2016.
Article in English | MEDLINE | ID: mdl-26703799

ABSTRACT

Lung cancer is the leading cause of cancer-related deaths worldwide with a 5-year overall survival rate of less than 20 %. Considering the treatments currently available, this statistics is shocking. A possible explanation for the disconnect between sophisticated treatments and the survival rate can be related to the post-treatment enrichment of Cancer Stem Cells (CSCs), which is one of a sub-set of drug resistant tumor cells with abilities of self-renewal, cancer initiation, and further maintenance of tumors. Lung CSCs have been associated with resistance to radiation and chemotherapeutic treatments. CSCs have also been implicated in tumor recurrence because CSCs are not typically killed after conventional therapy. Investigation of CSCs in determining their role in tumor recurrence and drug-resistance relied heavily on the use of specific markers present in CSCs, including CD133, ALDH, ABCG2, and Nanog. Yet another cell type that is also associated with increased resistance to treatment is epithelial-to-mesenchymal transition (EMT) phenotypic cells. Through the processes of EMT, epithelial cells lose their epithelial phenotype and gain mesenchymal properties, rendering EMT phenotypic cells acquire drug-resistance. In this chapter, we will further discuss the role of microRNAs (miRNAs) especially because miRNA-based therapies are becoming attractive target with respect to therapeutic resistance and CSCs. Finally, the potential role of the natural agents and synthetic derivatives of natural compounds with anti-cancer activity, e.g. curcumin, CDF, and BR-DIM is highlighted in overcoming therapeutic resistance, suggesting that the above mentioned agents could be important for better treatment of lung cancer in combination therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , Biomarkers, Tumor/genetics , Carcinoma, Non-Small-Cell Lung/drug therapy , Curcumin/therapeutic use , Lung Neoplasms/drug therapy , Neoplasm Recurrence, Local/drug therapy , Neoplastic Stem Cells/drug effects , Biomarkers, Tumor/metabolism , Carcinoma, Non-Small-Cell Lung/genetics , Carcinoma, Non-Small-Cell Lung/metabolism , Carcinoma, Non-Small-Cell Lung/pathology , Cell Proliferation/drug effects , Curcumin/analogs & derivatives , Curcumin/chemical synthesis , Drug Resistance, Neoplasm/genetics , Epithelial-Mesenchymal Transition/drug effects , Epithelial-Mesenchymal Transition/genetics , Gene Expression Regulation, Neoplastic , Humans , Lung Neoplasms/genetics , Lung Neoplasms/metabolism , Lung Neoplasms/pathology , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Neoplasm Recurrence, Local/genetics , Neoplasm Recurrence, Local/metabolism , Neoplasm Recurrence, Local/pathology , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Signal Transduction
3.
J Biol Chem ; 289(21): 14520-33, 2014 May 23.
Article in English | MEDLINE | ID: mdl-24719318

ABSTRACT

Subpopulations of cancer stem cells (CSCs) or cancer stem-like cells (CSLCs) have been identified from most tumors, including pancreatic cancer (PC), and the existence of these cells is clinically relevant. Emerging evidence suggests that CSLCs participate in cell growth/proliferation, migration/invasion, metastasis, and chemo-radiotherapy resistance, ultimately contributing to poor clinical outcome. However, the pathogenesis and biological significance of CSLCs in PC has not been well characterized. In the present study, we found that isolated triple-marker-positive (CD44(+)/CD133(+)/EpCAM(+)) cells of human PC MiaPaCa-2 and L3.6pl cells behave as CSLCs. These CSLCs exhibit aggressive behavior, such as increased cell growth, migration, clonogenicity, and self-renewal capacity. The mRNA expression profiling analysis showed that CSLCs (CD44(+)/CD133(+)/EpCAM(+)) exhibit differential expression of more than 1,600 mRNAs, including FoxQ1, compared with the triple-marker-negative (CD44(-)/CD133(-)/EpCAM(-)) cells. The knockdown of FoxQ1 by its siRNA in CSLCs resulted in the inhibition of aggressive behavior, consistent with the inhibition of EpCAM and Snail expression. Mouse xenograft tumor studies showed that CSLCs have a 100-fold higher potential for tumor formation and rapid tumor growth, consistent with overexpression of CSC-associated markers/mediators, including FoxQ1, compared with its parental MiaPaCa-2 cells. The inhibition of FoxQ1 attenuated tumor formation and growth, and expression of CSC markers in the xenograft tumor derived from CSLCs of MiaPaCa-2 cells. These data clearly suggest the role of differentially expressed genes in the regulation of CSLC characteristics, further suggesting that targeting some of these genes could be important for the development of novel therapies for achieving better treatment outcome of PC.


Subject(s)
Forkhead Transcription Factors/genetics , Neoplastic Stem Cells/metabolism , Pancreatic Neoplasms/genetics , AC133 Antigen , Animals , Antigens, CD/metabolism , Antigens, Neoplasm/metabolism , Apoptosis/genetics , Blotting, Western , Cell Adhesion Molecules/metabolism , Cell Cycle/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Proliferation , Epithelial Cell Adhesion Molecule , Forkhead Transcription Factors/metabolism , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Glycoproteins/metabolism , Humans , Hyaluronan Receptors/metabolism , Immunohistochemistry , Mice , Mice, SCID , Microscopy, Confocal , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Peptides/metabolism , RNA Interference , Reverse Transcriptase Polymerase Chain Reaction , Transcriptional Activation , Xenograft Model Antitumor Assays
4.
J Cell Physiol ; 229(3): 271-6, 2014 Mar.
Article in English | MEDLINE | ID: mdl-24037862

ABSTRACT

Prostate cancer (PCa) is the second leading cause of cancer death in men. Current research findings suggest that the androgen receptor (AR) and its signaling pathway contribute significantly to the progression of metastatic PCa. The AR is a ligand activated transcription factor, where androgens such as testosterone (T) and dihydroxytestosterone (DHT) act as the activating ligands. However in many metastatic PCa, the AR functions promiscuously and is constitutively active through multiple mechanisms. Inhibition of enzymes that take part in androgen synthesis or synthesizing antiandrogens that can inhibit the AR are two popular methods of impeding the androgen receptor signaling axis; however, the inhibition of androgen-independent activated AR function has not yet been fully exploited. This article focuses on the development of emerging novel agents that act at different steps along the androgen-AR signaling pathway to help improve the poor prognosis of PCa patients.


Subject(s)
Androgen Antagonists/therapeutic use , Antineoplastic Agents, Hormonal/therapeutic use , Neoplasms, Hormone-Dependent/drug therapy , Prostatic Neoplasms/drug therapy , Receptors, Androgen/drug effects , Androgen Antagonists/chemistry , Animals , Antineoplastic Agents, Hormonal/chemistry , Drug Design , Humans , Ligands , Male , Molecular Targeted Therapy , Neoplasms, Hormone-Dependent/metabolism , Neoplasms, Hormone-Dependent/pathology , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Receptors, Androgen/metabolism , Signal Transduction/drug effects , Treatment Outcome
5.
J Cancer Res Ther ; 20(1): 457-459, 2024 Jan 01.
Article in English | MEDLINE | ID: mdl-38554363

ABSTRACT

Renal cell carcinomas are known for their unforeseeable metastatic pattern. They are known to have high metastatic potential, thus commonly associated with synchronous or metachronous metastatic presentation. At the time of diagnosis, approximately one-third of patients present with metastatic disease. We present a case of synchronous metastasis of clear cell carcinoma to the gallbladder in a 54-year-old male within two months after radical nephrectomy.


Subject(s)
Carcinoma, Renal Cell , Kidney Neoplasms , Humans , Male , Middle Aged , Carcinoma, Renal Cell/diagnosis , Carcinoma, Renal Cell/surgery , Carcinoma, Renal Cell/pathology , Kidney Neoplasms/pathology , Nephrectomy
6.
Biochim Biophys Acta ; 1825(2): 160-72, 2012 Apr.
Article in English | MEDLINE | ID: mdl-22155217

ABSTRACT

Over decades, cancer treatment has been mainly focused on targeting cancer cells and not much attention to host tumor microenvironment. Recent advances suggest that the tumor microenvironment requires in-depth investigation for understanding the interactions between tumor cell biology and immunobiology in order to optimize therapeutic approaches. Tumor microenvironment consists of cancer cells and tumor associated reactive fibroblasts, infiltrating non-cancer cells, secreted soluble factors or molecules, and non-cellular support materials. Tumor associated host immune cells such as Th(1), Th(2), Th17, regulatory cells, dendritic cells, macrophages, and myeloid-derived suppressor cells are major components of the tumor microenvironment. Accumulating evidence suggests that these tumor associated immune cells may play important roles in cancer development and progression. However, the exact functions of these cells in the tumor microenvironment are poorly understood. In the tumor microenvironment, NF-κB plays an important role in cancer development and progression because this is a major transcription factor which regulates immune functions within the tumor microenvironment. In this review, we will focus our discussion on the immunological contribution of NF-κB in tumor associated host immune cells within the tumor microenvironment. We will also discuss the potential protective role of zinc, a well-known immune response mediator, in the regulation of these immune cells and cancer cells in the tumor microenvironment especially because zinc could be useful for conditioning the tumor microenvironment toward innovative cancer therapy.


Subject(s)
Antineoplastic Agents/therapeutic use , NF-kappa B/immunology , Neoplasms/drug therapy , Neoplasms/immunology , Tumor Microenvironment/immunology , Zinc/therapeutic use , Animals , Cell Line, Tumor , Disease Progression , Humans
7.
Biochim Biophys Acta ; 1826(2): 272-96, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22579961

ABSTRACT

Hypoxia is one of the fundamental biological phenomena that are intricately associated with the development and aggressiveness of a variety of solid tumors. Hypoxia-inducible factors (HIF) function as a master transcription factor, which regulates hypoxia responsive genes and has been recognized to play critical roles in tumor invasion, metastasis, and chemo-radiation resistance, and contributes to increased cell proliferation, survival, angiogenesis and metastasis. Therefore, tumor hypoxia with deregulated expression of HIF and its biological consequence lead to poor prognosis of patients diagnosed with solid tumors, resulting in higher mortality, suggesting that understanding of the molecular relationship of hypoxia with other cellular features of tumor aggressiveness would be invaluable for developing newer targeted therapy for solid tumors. It has been well recognized that cancer stem cells (CSCs) and epithelial-to-mesenchymal transition (EMT) phenotypic cells are associated with therapeutic resistance and contribute to aggressive tumor growth, invasion, metastasis and believed to be the cause of tumor recurrence. Interestingly, hypoxia and HIF signaling pathway are known to play an important role in the regulation and sustenance of CSCs and EMT phenotype. However, the molecular relationship between HIF signaling pathway with the biology of CSCs and EMT remains unclear although NF-κB, PI3K/Akt/mTOR, Notch, Wnt/ß-catenin, and Hedgehog signaling pathways have been recognized as important regulators of CSCs and EMT. In this article, we will discuss the state of our knowledge on the role of HIF-hypoxia signaling pathway and its kinship with CSCs and EMT within the tumor microenvironment. We will also discuss the potential role of hypoxia-induced microRNAs (miRNAs) in tumor development and aggressiveness, and finally discuss the potential effects of nutraceuticals on the biology of CSCs and EMT in the context of tumor hypoxia.


Subject(s)
Cell Hypoxia , Epithelial-Mesenchymal Transition , MicroRNAs/physiology , Neoplasms/pathology , Neoplastic Stem Cells/physiology , Animals , Apoptosis Regulatory Proteins , Basic Helix-Loop-Helix Transcription Factors/physiology , Gene Expression Regulation, Neoplastic , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/physiology , NF-kappa B/physiology , Prognosis , Repressor Proteins , Signal Transduction , Vascular Endothelial Growth Factor A/physiology
8.
J Cell Physiol ; 228(1): 36-42, 2013 Jan.
Article in English | MEDLINE | ID: mdl-22689345

ABSTRACT

Researchers and clinicians have been challenged with the development of therapies for the treatment of cancer patients whose tumors metastasized to the brain. Among the most lethal weapons known today, current management of brain metastases involves multiple therapeutic modalities that provide little, if any, for improving the quality of life and overall survival. Recently the role of cancer stem cells (CSCs) in the development of cancer has been studied extensively, and thus its role in the prognosis, diagnosis, and treatment is now being investigated even in the realm of brain metastasis (BM). Recognizing the molecular make-up of CSCs as well as understanding the role of these cells in resistance to treatment modalities is expected to benefit cancer patients. Additionally, past decade has witnessed an increase in awareness and understanding of the role of microRNAs (miRNAs) in various cancer types, and the deregulation miRNAs are critically important for the regulation of genes during the development and progression of human malignancies. The role miRNAs in BM is being investigated, and has also shown tremendous promise for future research. In this review, we discuss the problem and lethality of brain metastases and the current state of management, and further provide insight into novel avenues that are worth considering including the biological complexities of CSCs and miRNAs for designing novel therapies.


Subject(s)
Brain Neoplasms/pathology , MicroRNAs/metabolism , Neoplastic Stem Cells/metabolism , Antineoplastic Agents/administration & dosage , Antineoplastic Agents/therapeutic use , Brain Neoplasms/drug therapy , Brain Neoplasms/secondary , Humans , Signal Transduction
9.
J Cell Physiol ; 228(3): 556-562, 2013 Mar.
Article in English | MEDLINE | ID: mdl-22806240

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is one of the most frequently diagnosed cancers and the fourth leading cause of cancer-related death in the United States, suggesting that there is an urgent need to design novel strategies for achieving better treatment outcome of patients diagnosed with PDAC. Our previous study has shown that activation of Notch and NF-κB play a critical role in the development of PDAC in the compound K-Ras(G12D) and Ink4a/Arf deficient transgenic mice. However, the exact molecular mechanism by which mutated K-Ras and Ink4a/Arf deficiency contribute to progression of PDAC remains largely elusive. In the present study, we used multiple methods, such as real-time RT-PCR, Western blotting assay, and immunohistochemistry to gain further mechanistic insight. We found that the deletion of Ink4a/Arf in K-Ras(G12D) expressing mice led to high expression of PDGF-D signaling pathway in the tumor and tumor-derived cell line (RInk-1 cells). Furthermore, PDGF-D knock-down in RInk-1 cells resulted in the inhibition of pancreatosphere formation and down-regulation of EZH2, CD44, EpCAM, and vimentin. Moreover, we demonstrated that epithelial-mesenchymal transition (EMT) was induced in the compound mice, which is linked with aggressiveness of PDAC. In addition, we demonstrated that tumors from compound transgenic mice have higher expression of cancer stem cell (CSC) markers. These results suggest that the acquisition of EMT phenotype and induction of CSC characteristics could be linked with the aggressiveness of PDAC mediated in part through the activation of PDGF-D, signaling.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/deficiency , Pancreatic Neoplasms/etiology , Proto-Oncogene Proteins p21(ras)/deficiency , Animals , Carcinoma, Pancreatic Ductal/etiology , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cyclin-Dependent Kinase Inhibitor p16/genetics , Disease Progression , Epithelial-Mesenchymal Transition , Genes, ras , Humans , Lymphokines/deficiency , Lymphokines/genetics , Mice , Mice, Transgenic , Mutation , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Pancreatic Neoplasms/pathology , Phenotype , Platelet-Derived Growth Factor/deficiency , Platelet-Derived Growth Factor/genetics , Proto-Oncogene Proteins p21(ras)/genetics , Signal Transduction
10.
Int J Mol Sci ; 14(7): 14771-84, 2013 Jul 16.
Article in English | MEDLINE | ID: mdl-23863689

ABSTRACT

Cancer diagnosis is currently undergoing a paradigm shift with the incorporation of molecular biomarkers as part of routine diagnostic panel. The molecular alteration ranges from those involving the DNA, RNA, microRNAs (miRNAs) and proteins. The miRNAs are recently discovered small non-coding endogenous single-stranded RNAs that critically regulates the development, invasion and metastasis of cancers. They are altered in cancers and have the potential to serve as diagnostic markers for cancer. Moreover, deregulating their activity offers novel cancer therapeutic approaches. The availability of high throughput techniques for the identification of altered cellular molecules allowed their use in cancer diagnosis. Their application to a variety of body specimens from blood to tissues has been helpful for appreciating their use in the clinical context. The development of innovative antibodies for immunohistochemical detection of proteins also assists in diagnosis and risk stratification. Overall, the novel cancer diagnostic tools have extended their application as prognostic risk factors and can be used as targets for personalized medicine.


Subject(s)
Biomarkers/metabolism , Neoplasms/metabolism , DNA/metabolism , Humans , MicroRNAs/metabolism , Neoplasms/diagnosis , Neoplasms/drug therapy , Proteome/metabolism , RNA/metabolism
11.
Obes Surg ; 33(6): 1694-1701, 2023 06.
Article in English | MEDLINE | ID: mdl-37087518

ABSTRACT

PURPOSE: The clinical benefits of bariatric surgery are well-established, but the impact of bariatric surgery on psychosocial outcomes such as health-related quality of life (HRQL) is less clear. The aim of this study is to assess the Quality of life (QOL) as a whole and in separate domains in post-bariatric surgery patients. METHODOLOGY: A single unit cross-sectional analysis of a prospective study is done on QOL in 51 patients undergoing laparoscopic sleeve gastrectomy at tertiary hospital. QOL was assessed by WHOQOL-BREF (World Health Organisation Quality of Life questionnaire -Brief version) questionnaire and Global Quality of Life Scale in each patient. Scores were calculated on a 0-100 scale and results compared. RESULTS: The median scores given by patients before surgery were 14, 21, 42, 40 and 12.5 for each of the parameters physical, psychological, social, environmental and overall well-being respectively. The median scores for after surgery were 86, 87, 91, 88 and 87.5 respectively. The difference was significant (p value 0.001). Global QOL after surgery, calculated year wise, showed QOL scores of 90, 100, 95 and 80 in patients with 1 year, 2 years, 3 years and 4 years of follow-up without any significant difference (p value 0.502). CONCLUSION: Through this study, we emphasize the need for the selection of a standardised scale by international organisations to compare the different studies. By proving the significant differences in the QOL of patients who underwent LSG [laparoscopic sleeve gastrectomy], we suggest to consider the Quality of Life as one of the criteria to consider a patient for bariatric surgery.


Subject(s)
Bariatric Surgery , Laparoscopy , Obesity, Morbid , Humans , Quality of Life , Obesity, Morbid/surgery , Prospective Studies , Cross-Sectional Studies , Weight Loss , Bariatric Surgery/methods , Gastrectomy/methods , Laparoscopy/methods , Treatment Outcome
12.
Carcinogenesis ; 33(8): 1563-71, 2012 Aug.
Article in English | MEDLINE | ID: mdl-22637745

ABSTRACT

Membrane type-1 matrix metalloproteinase (MT1-MMP) is often activated and expressed in tumor cells with significant invasive properties, and is associated with poor prognosis of patients. This could partly be due to deregulated expression of microRNAs (miRNAs) which regulates the expression of MT1-MMP and PTEN (phosphatase and tensin homolog) contributing to tumor invasion and metastasis. We initially compared the expression profile of miR-200 family, PTEN and MT1-MMP expression in six pancreatic cancer (PC) cell lines by qRT-PCR and western blot analysis. We found loss of expression of miR-200a, b and c in chemo-resistant PC cell lines, which was correlated with loss of PTEN and over-expression of MT1-MMP. Based on our initial findings, we chose BxPC-3, MIAPaCa-2 and MIAPaCa-2-GR cells for further mechanistic studies We assessed the effect of two separate novel agents CDF (a synthetic analog of curcumin) and BR-DIM (a natural agent) on PC cells. The expression of miR-200 family and PTEN was significantly re-expressed whereas the expression of MT1-MMP was down-regulated by CDF and BR-DIM treatment. Forced over-expression or silencing of miR-200c, followed by either CDF or BR-DIM treatment of MIAPaCa-2 cells, altered the morphology of cells, wound-healing capacity, colony formation and the expression of MT1-MMP and PTEN. These results provide strong experimental evidence showing that the loss of miR-200 family and PTEN expression and increased level of MT1-MMP leads to aggressive behavior of PC cells, which could be attenuated through re-expression of miR-200c by CDF and/or BR-DIM treatment, suggesting that these agents could be useful for PC treatment.


Subject(s)
Matrix Metalloproteinase 1/metabolism , MicroRNAs/metabolism , PTEN Phosphohydrolase/metabolism , Pancreatic Neoplasms/metabolism , Humans , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology
13.
Carcinogenesis ; 33(12): 2450-6, 2012 Dec.
Article in English | MEDLINE | ID: mdl-22971573

ABSTRACT

Garcinol, obtained from Garcinia indica, has exhibited some promising anticancer activity. In particular, our earlier work has demonstrated its ability to inhibit cell proliferation and induction of apoptosis in multiple cancer cell lines representative of breast, prostate, as well as pancreatic cancers. However, its exact mechanism of action remains largely unclear. Here we show that garcinol also targets signal transducer and activator of transcription-3 (STAT-3) signaling pathway. STAT-3 is frequently found to be activated in many cancer types and this is the first report on such action of garcinol leading to its anticancer effects. Garcinol inhibited total, as well as phosphorylated, STAT-3 in breast, prostate and pancreatic cancer cell lines and was also found to inhibit cell invasion of all the cancer cell lines tested. STAT-3 phosphorylation was inhibited by garcinol in a dose-dependent manner. We also observed an inhibitory effect of garcinol on IL-6-induced STAT-3 phosphorylation and production of urokinase-type plasminogen activator, vascular endothelial growth factor and matrix metalloproteinase-9, which might explain the reduced invasion and aggressiveness of cells treated with garcinol. The results were further verified in vivo using MDA-MB-231 breast cancer mouse xenograft model where administration of garcinol significantly inhibited tumor growth, and western blot analysis of remnant tumor lysates showed reduced STAT-3 expression and activation. These results suggest that garcinol may have translational potential as chemopreventive or therapeutic agent against multiple cancers and inhibition of STAT-3 signaling pathway is one of the mechanisms by which garcinol exerts its anticancer effects.


Subject(s)
Antineoplastic Agents/pharmacology , STAT3 Transcription Factor/antagonists & inhibitors , Signal Transduction/drug effects , Terpenes/pharmacology , Animals , Cell Line, Tumor , Chlorine Compounds/pharmacology , Female , Humans , Interleukin-6/pharmacology , Mice , Mice, Inbred ICR , NF-kappa B/physiology , Neoplasm Invasiveness , Platinum Compounds/pharmacology , STAT3 Transcription Factor/genetics , STAT3 Transcription Factor/physiology
14.
Biochim Biophys Acta ; 1815(2): 135-46, 2011 Apr.
Article in English | MEDLINE | ID: mdl-21129444

ABSTRACT

Pancreatic cancer (PC) is one of the most lethal malignant diseases with the worst prognosis. It is ranked as the fourth leading cause of cancer-related deaths in the United States. Many risk factors have been associated with PC. Interestingly, large numbers of epidemiological studies suggest that obesity and diabetes, especially type-2 diabetes, are positively associated with increased risk of PC. Similarly, these chronic diseases (obesity, diabetes, and cancer) are also a major public health concern. In the U.S. population, 50 percent are overweight, 30 percent are medically obese, and 10 percent have diabetes mellitus (DM). Therefore, obesity and DM have been considered as potential risk factors for cancers; however, the focus of this article is restricted to PC. Although the mechanisms responsible for the development of these chronic diseases leading to the development of PC are not fully understood, the biological importance of the activation of insulin, insulin like growth factor-1 (IGF-1) and its receptor (IGF-1R) signaling pathways in insulin resistance mechanism and subsequent induction of compensatory hyperinsulinemia has been proposed. Therefore, targeting insulin/IGF-1 signaling with anti-diabetic drugs for lowering blood insulin levels and reversal of insulin resistance could be useful strategy for the prevention and/or treatment of PC. A large number of studies have demonstrated that the administration of anti-diabetic drugs such as metformin and thiazolidinediones (TZD) class of PPAR-γ agonists decreases the risk of cancers, suggesting that these agents might be useful anti-tumor agents for the treatment of PC. In this review article, we will discuss the potential roles of metformin and TZD anti-diabetic drugs as anti-tumor agents in the context of PC and will further discuss the complexities and the possible roles of microRNAs (miRNAs) in the pathogenesis of obesity, diabetes, and PC.


Subject(s)
Diabetes Complications , Obesity/complications , Pancreatic Neoplasms/etiology , Disease Progression , Humans , Insulin Resistance , Pancreatic Neoplasms/physiopathology , Prognosis , Risk Factors
15.
J Cell Physiol ; 227(10): 3373-80, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22213426

ABSTRACT

Human pancreatic cancer (PC) is an aggressive disease, which has been recapitulated in transgenic animal model that provides unique opportunity for mechanistic understanding of disease progression and also for testing the efficacy of novel therapeutics. Emerging evidence suggests deregulated expression of microRNAs (miRNAs) in human PC, and thus we investigated the expression of miRNAs in pancreas tissues obtained from transgenic mouse models of K-Ras (K), Pdx1-Cre (C), K-Ras;Pdx1-Cre (KC), and K-Ras;Pdx1-Cre;INK4a/Arf (KCI), initially from pooled RNA samples using miRNA profiling, and further confirmed in individual specimens by quantitative RT-PCR. We found over-expression of miR-21, miR-221, miR-27a, miR-27b, and miR-155, and down-regulation of miR-216a, miR-216b, miR-217, and miR-146a expression in tumors derived from KC and KCI mouse model, which was consistent with data from KCI-derived RInk-1 cells. Mechanistic investigations revealed a significant induction of EGFR, K-Ras, and MT1-MMP protein expression in tissues from both KC and KCI mouse compared to tissues from K or C, and these results were consistent with similar findings in RInk-1 cells compared to human MIAPaCa-2 cells. Furthermore, miR-155 knock-down in RInk-1 cells resulted in the inhibition of cell growth and colony formation consistent with down-regulation of EGFR, MT1-MMP, and K-Ras expression. In addition, miR-216b which target Ras, and forced re-expression of miR-216b in RInk-1 cells showed inhibition of cell proliferation and colony formation, which was correlated with reduced expression of Ras, EGFR, and MT1-MMP. These findings suggest that these models would be useful for preclinical evaluation of novel miRNA-targeted agents for designing personalized therapy for PC.


Subject(s)
Cyclin-Dependent Kinase Inhibitor p16/metabolism , Down-Regulation/genetics , Genes, ras , MicroRNAs/genetics , Pancreatic Neoplasms/genetics , Animals , Cell Line , Cell Proliferation , Cyclin-Dependent Kinase Inhibitor p16/genetics , ErbB Receptors/genetics , ErbB Receptors/metabolism , Gene Expression Regulation, Neoplastic/genetics , Humans , Matrix Metalloproteinase 14/genetics , Matrix Metalloproteinase 14/metabolism , Mice , Mice, Transgenic/genetics , Mice, Transgenic/metabolism , MicroRNAs/metabolism , Pancreas/metabolism , Pancreatic Neoplasms/metabolism , Stem Cells/metabolism
16.
Int J Mol Sci ; 13(10): 12556-72, 2012 Oct 01.
Article in English | MEDLINE | ID: mdl-23202913

ABSTRACT

Cancer epidemiology and prevention is one of the most well studied fields today. The more we can understand about the incidence and pathogenesis of this disease, the better we will be able to prevent it. Effective prevention strategies can decrease the mortality rate of cancer significantly; this is why it is important to delineate the underlying causes. It has been well recognized that genetic mutations, sporadic or hereditary, may lead to increased chance of tumorigenesis. Detecting genetic mutations can lead to the identification of high-risk individuals with hereditary cancer syndromes, which may assist in devising prevention strategies. Further, environmental factors are known to play important roles in epidemiology and suggest prevention tools that could be implemented to reduce cancer incidence and subsequent cancer-associated morbidity and mortality. Chemoprevention has been tried in colon cancer and is finding new advancements in other carcinomas as well. Out of many environmental cancer preventive agents, the most notable developments are the identification of the role of vitamins E, vitamin D and folic acid. Increased consumption of these vitamins has shown to be inversely correlated with cancer risk. This review will highlight important aspects of cancer epidemiology in the most aggressive carcinomas of the gastrointestinal system focusing on colorectal adenocarcinoma and pancreatic adenocarcinoma. Additionally, some of the well-known and evolving aspects of epidemiology of colorectal and pancreatic cancer along with current and new prevention strategies will also be reviewed.


Subject(s)
Gastrointestinal Neoplasms/epidemiology , Colorectal Neoplasms/epidemiology , Colorectal Neoplasms/genetics , Colorectal Neoplasms/prevention & control , Cyclooxygenase 2 Inhibitors/therapeutic use , Diabetes Complications/pathology , Folic Acid/metabolism , Gastrointestinal Neoplasms/prevention & control , Humans , Hypoglycemic Agents/therapeutic use , Inflammatory Bowel Diseases/complications , Inflammatory Bowel Diseases/pathology , Metformin/therapeutic use , Obesity/complications , Obesity/pathology , Pancreatic Neoplasms/epidemiology , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/prevention & control , Smoking
17.
J Cell Biochem ; 112(9): 2296-306, 2011 Sep.
Article in English | MEDLINE | ID: mdl-21503965

ABSTRACT

FoxM1 is known to play important role in the development and progression of many malignancies including pancreatic cancer. Studies have shown that the acquisition of epithelial-to-mesenchymal transition (EMT) phenotype and induction of cancer stem cell (CSC) or cancer stem-like cell phenotypes are highly inter-related, and contributes to drug resistance, tumor recurrence, and metastasis. The molecular mechanism(s) by which FoxM1 contributes to the acquisition of EMT phenotype and induction of CSC self-renewal capacity is poorly understood. Therefore, we established FoxM1 over-expressing pancreatic cancer (AsPC-1) cells, which showed increased cell growth, clonogenicity, and cell migration. Moreover, over-expression of FoxM1 led to the acquisition of EMT phenotype by activation of mesenchymal cell markers, ZEB1, ZEB2, Snail2, E-cadherin, and vimentin, which is consistent with increased sphere-forming (pancreatospheres) capacity and expression of CSC surface markers (CD44 and EpCAM). We also found that over-expression of FoxM1 led to decreased expression of miRNAs (let-7a, let-7b, let-7c, miR-200b, and miR-200c); however, re-expression of miR-200b inhibited the expression of ZEB1, ZEB2, vimentin as well as FoxM1, and induced the expression of E-cadherin, leading to the reversal of EMT phenotype. Finally, we found that genistein, a natural chemo-preventive agent, inhibited cell growth, clonogenicity, cell migration and invasion, EMT phenotype, and formation of pancreatospheres consistent with reduced expression of CD44 and EpCAM. These results suggest, for the first time, that FoxM1 over-expression is responsible for the acquisition of EMT and CSC phenotype, which is in part mediated through the regulation of miR-200b and these processes, could be easily attenuated by genistein.


Subject(s)
Epithelial-Mesenchymal Transition , Forkhead Transcription Factors/metabolism , Neoplastic Stem Cells/metabolism , Anticarcinogenic Agents/pharmacology , Antigens, Differentiation/genetics , Antigens, Differentiation/metabolism , Cell Aggregation/drug effects , Cell Line, Tumor , Cell Movement/drug effects , Cell Proliferation , Cell Shape , Cell Survival/drug effects , Forkhead Box Protein M1 , Forkhead Transcription Factors/genetics , Gene Expression Profiling , Gene Expression Regulation, Neoplastic , Genetic Markers , Genistein/pharmacology , Humans , MicroRNAs/genetics , MicroRNAs/metabolism , Neoplastic Stem Cells/pathology , Pancreatic Neoplasms
18.
Int J Cancer ; 129(7): 1781-91, 2011 Oct 01.
Article in English | MEDLINE | ID: mdl-21154750

ABSTRACT

Emerging evidence suggests that the transcription factor Forkhead Box M1 (FoxM1) is associated with aggressive human carcinomas, including breast cancer. Because elevated expression of FoxM1 has been observed in human breast cancers, FoxM1 has attracted much attention in recent years as a potential target for the prevention and/or therapeutic intervention in breast cancer. However, no information is currently available regarding how downregulation of FoxM1 could be achieved for breast cancer prevention and therapy. Here, we report for the first time that 3,3'-diindolylmethane (DIM), a nontoxic dietary chemopreventive agent could effectively downregulate FoxM1 in various breast cancer cell lines. Using gene transfection, real-time reverse transcription-PCR, Western blotting, invasion and 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide assays, we found that DIM could enhance Taxotere-induced growth inhibition of breast cancer cells, and decreased invasive capacity of breast cancer cells was observed after either treatment alone or the combination. These effects were associated with downregulation of FoxM1. We also found that knock down of FoxM1 expression by small interfering RNA (siRNA) transfection increased DIM-induced cell growth inhibition, whereas over-expression of FoxM1 by cDNA transfection attenuated DIM-induced cell growth inhibition, suggesting the mechanistic role of FoxM1. Most importantly, the combination treatment significantly inhibited tumor growth in severe combined immunodeficiency (SCID) mice, and the results were correlated with the downregulation of FoxM1 in tumor remnants. We conclude that inactivation of FoxM1 and its target genes by DIM could enhance the therapeutic efficacy of Taxotere in breast cancer, which could be a useful strategy for the prevention and/or treatment of breast cancer.


Subject(s)
Breast Neoplasms/drug therapy , Breast Neoplasms/genetics , Forkhead Transcription Factors/genetics , Indoles/pharmacology , Taxoids/pharmacology , Animals , Cell Cycle/drug effects , Cell Line, Tumor , Cell Proliferation/drug effects , Docetaxel , Down-Regulation , Forkhead Box Protein M1 , Forkhead Transcription Factors/metabolism , Humans , Mice , Mice, Nude , Xenograft Model Antitumor Assays
19.
Biochem Biophys Res Commun ; 407(4): 703-7, 2011 Apr 22.
Article in English | MEDLINE | ID: mdl-21439265

ABSTRACT

Zinc deficiency impairs cellular immunity. Up-regulation of mRNA levels of IFN-γ, IL-12Rß2, and T-bet are essential for Th(1) differentiation. We hypothesized that zinc increases Th(1) differentiation via up-regulation of IFN-γ and T-bet expression. To test this hypothesis, we used zinc-deficient and zinc-sufficient HUT-78 cells (a Th(0) cell line) under different condition of stimulation in this study. We also used TPEN, a zinc-specific chelator, to decrease the bioavailability of zinc in the cells. We measured intracellular free zinc, cytokines, and the mRNAs of T-bet, IFN-γ, and IL-12Rß2. In this study, we show that in zinc-sufficient HUT-78 cells, mRNA levels of IFN-γ, IL-12Rß2, and T-bet in PMA/PHA-stimulated cells were increased in comparison to zinc-deficient cells. Although intracellular free zinc was increased slightly in PMA/PHA-stimulated cells, Con-A-stimulated cells in 5µM zinc medium showed a greater sustained increase in intracellular free zinc in comparison to cells incubated in 1µM zinc. The cells pre-incubated with TPEN showed decreased mRNA levels of IFN-γ and T-bet mRNAs in comparison to cells without TPEN incubation. We conclude that stimulation of cells by Con-A via TCR, release intracellular free zinc which functions as a signal molecule for generation of IFN-γ and T-bet, and IL-12Rß2 mRNAs required for Th(1) cell differentiation. These results suggest that zinc increase Th(1) cell differentiation by up-regulation of IFN-γ and T-bet, and IL-12Rbß2 mRNAs.


Subject(s)
Cell Differentiation , Interferon-gamma/biosynthesis , Receptors, Interleukin-12/biosynthesis , T-Box Domain Proteins/biosynthesis , Th1 Cells/immunology , Zinc/metabolism , Cell Differentiation/drug effects , Cell Differentiation/genetics , Cell Line , Concanavalin A/pharmacology , Humans , Interferon-gamma/genetics , Receptors, Interleukin-12/genetics , T-Box Domain Proteins/genetics , Th1 Cells/cytology , Up-Regulation , Zinc/deficiency , Zinc/pharmacology
SELECTION OF CITATIONS
SEARCH DETAIL