Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 18 de 18
Filter
Add more filters










Publication year range
1.
Cancers (Basel) ; 14(11)2022 Jun 06.
Article in English | MEDLINE | ID: mdl-35681792

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) is currently the third leading cause of cancer-related death in the United States. Even though the poor prognosis of PDAC is often attributed to late diagnosis, patients with an early diagnosis who undergo tumor resection and adjuvant chemotherapy still show tumor recurrence, highlighting a need to develop therapies which can overcome chemoresistance. Chemoresistance has been linked to the high expression of microRNAs (miRs), such as miR-21, within tumor cells. Tumor cells can collect miRs through the uptake of miR-containing lipid extracellular vesicles called exosomes. These exosomes are secreted in high numbers from cancer-associated fibroblasts (CAFs) within the tumor microenvironment during gemcitabine treatment and can contribute to cell proliferation and chemoresistance. Here, we show a novel mechanism in which CAF-derived exosomes may promote proliferation and chemoresistance, in part, through suppression of the tumor suppressor PTEN. We identified five microRNAs: miR-21, miR-181a, miR-221, miR-222, and miR-92a, that significantly increased in number within the CAF exosomes secreted during gemcitabine treatment which target PTEN. Furthermore, we found that CAF exosomes suppressed PTEN expression in vitro and that treatment with the exosome inhibitor GW4869 blocked PTEN suppression in vivo. Collectively, these findings highlight a mechanism through which the PTEN expression loss, often seen in PDAC, may be attained and lend support to investigations into the use of exosome inhibitors as potential therapeutics to improve the effectiveness of chemotherapy.

2.
Matrix Biol Plus ; 14: 100111, 2022 Jun.
Article in English | MEDLINE | ID: mdl-35619988

ABSTRACT

In pancreatic ductal adenocarcinoma (PDAC), the abundant stromal cells which comprise the tumor microenvironment constitute more than 90% of the primary tumor bulk. Moreover, this desmoplastic environment has been found to be three times stiffer than normal pancreas tissue. Despite the importance of studying the desmoplastic environment of PDAC, there is still a lack of models designed to adequately recapitulate this complex stiff microenvironment, a critical hallmark of the disease that has been shown to induce chemoresistance. Here, we present a bio-mimetic, 3-dimensional co-culture system that integrates tumor organoids and host-matching stromal cancer associated-fibroblasts (CAFs) that recapitulates the complex, fibrotic matrix of PDAC using advanced biomaterials. With this model, we show that matrix-activated CAFs are able to "re-engineer" the fibrotic environment into a significantly stiffer environment through lysyl-oxidase dependent crosslinking. Moreover, we show that culture of CAFs in this model leads to an increase of exosomes; extracellular vesicles known to promote chemoresistance. Finally, using previously identified exosome inhibitors, climbazole and imipramine, we demonstrate how abrogation of exosome hypersecretion can reduce matrix stiffness-induced chemoresistance. These data highlight the importance of the development of new models that recapitulate not only the cellular composition found in PDAC tumors, but also the biophysical stresses, like stiffness, that the cells are exposed to in order to identify therapies that can overcome this critical feature which can contribute to the chemoresistance observed in patients. We believe that the 3D bio-mimetic model we have developed will be a valuable tool for the development, testing, and optimization of "mechano-medicines" designed to target the biophysical forces that lead to tumor growth and chemoresistance.

3.
Commun Biol ; 2: 189, 2019.
Article in English | MEDLINE | ID: mdl-31123713

ABSTRACT

Extracellular vesicles (EV) containing microRNAs (miRNAs) have tremendous potential as biomarkers for the early detection of disease. Here, we present a simple and rapid PCR-free integrated microfluidics platform capable of absolute quantification (<10% uncertainty) of both free-floating miRNAs and EV-miRNAs in plasma with 1 pM detection sensitivity. The assay time is only 30 minutes as opposed to 13 h and requires only ~20 µL of sample as oppose to 1 mL for conventional RT-qPCR techniques. The platform integrates a surface acoustic wave (SAW) EV lysing microfluidic chip with a concentration and sensing microfluidic chip incorporating an electrokinetic membrane sensor that is based on non-equilibrium ionic currents. Unlike conventional RT-qPCR methods, this technology does not require EV extraction, RNA purification, reverse transcription, or amplification. This platform can be easily extended for other RNA and DNA targets of interest, thus providing a viable screening tool for early disease diagnosis, prognosis, and monitoring of therapeutic response.


Subject(s)
Extracellular Vesicles/chemistry , Lab-On-A-Chip Devices , MicroRNAs/blood , Animals , Biomarkers/blood , Equipment Design , Humans , Liver Neoplasms/blood , Liver Neoplasms/genetics , Male , Mice , Mice, Inbred C57BL , Mice, Knockout , MicroRNAs/genetics , Real-Time Polymerase Chain Reaction
4.
Electrophoresis ; 2018 Feb 27.
Article in English | MEDLINE | ID: mdl-29484678

ABSTRACT

Exosomes carry microRNA biomarkers, occur in higher abundance in cancerous patients than in healthy ones, and because they are present in most biofluids, including blood and urine, these can be obtained noninvasively. Standard laboratory techniques to isolate exosomes are expensive, time consuming, provide poor purity, and recover on the order of 25% of the available exosomes. We present a new microfluidic technique to simultaneously isolate exosomes and preconcentrate them by electrophoresis using a high transverse local electric field generated by ion-depleting ion-selective membrane. We use pressure-driven flow to deliver an exosome sample to a microfluidic chip such that the transverse electric field forces them out of the cross flow and into an agarose gel which filters out unwanted cellular debris while the ion-selective membrane concentrates the exosomes through an enrichment effect. We efficiently isolated exosomes from 1× PBS buffer, cell culture media, and blood serum. Using flow rates from 150 to 200 µL/h and field strengths of 100 V/cm, we consistently captured between 60 and 80% of exosomes from buffer, cell culture media, and blood serum as confirmed by both fluorescence spectroscopy and nanoparticle tracking analysis. Our microfluidic chip maintained this recovery rate for more than 20 min with a concentration factor of 15 for 10 min of isolation.

5.
Neoplasia ; 20(1): 12-24, 2018 01.
Article in English | MEDLINE | ID: mdl-29175021

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) presents at metastatic stage in over 50% of patients. With a survival rate of just 2.7% for patients presenting with distant disease, it is imperative to uncover novel mechanisms capable of suppressing metastasis in PDAC. Previously, we reported that the loss of metastasis suppressor protein 1 (MTSS1) in PDAC cells results in significant increase in cellular migration and invasion. Conversely, we also found that overexpressing MTSS1 in metastatic PDAC cell lines corresponds with not only decreased metastatic phenotype, but also greater overall survival. While it is known that MTSS1 is downregulated in late-stage PDAC, the mechanism behind that loss has not yet been elucidated. Here, we build off our previous findings to present a novel regulatory mechanism for the stabilization of MTSS1 via the tumor suppressor protein phosphatase and tensin homolog (PTEN). We show that PTEN loss in PDAC cells results in a decrease in MTSS1 expression and increased metastatic potential. Additionally, we demonstrate that PTEN forms a complex with MTSS1 in order to stabilize and protect it from proteasomal degradation. Finally, we show that the inflammatory tumor microenvironment, which makes up over 90% of PDAC tumor bulk, is capable of downregulating PTEN expression through secretion of miRNA-23b, potentially uncovering a novel extrinsic mechanism of MTSS1 regulation. Collectively, these data offer new insight into the role and regulation of MTSS1in suppressing tumor cell invasion and migration and help shed light as to what molecular mechanisms could be leading to early cell dissemination in PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Microfilament Proteins/metabolism , Neoplasm Proteins/metabolism , PTEN Phosphohydrolase/genetics , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Animals , Carcinoma, Pancreatic Ductal/mortality , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Cell Movement/genetics , Gene Expression , Humans , Mice , MicroRNAs , Neoplasm Metastasis , PTEN Phosphohydrolase/metabolism , Pancreatic Neoplasms/mortality , Pancreatic Neoplasms/pathology , Phenotype , Prognosis , Proteasome Endopeptidase Complex/metabolism , Protein Binding , Protein Stability , Proteolysis
6.
Curr Drug Targets ; 19(6): 701-708, 2018.
Article in English | MEDLINE | ID: mdl-28641518

ABSTRACT

BACKGROUND: Resistance to therapy is a major hindrance to patient survival in cancer, underscoring a critical need to elucidate the underlying mechanisms responsible for chemoresistance. Research has demonstrated that endoplasmic reticulum (ER) stress plays a significant role in allowing cells to survive conditions that would normally elicit cell death. Specifically, elevated expression of GRP78, the master regulator of the unfolded protein response (UPR), has been shown to induce chemoresistance and serves as a indictor of poor prognosis in patients. OBJECTIVE: Evaluating the expression of GRP78 and its downstream targets in a wide range of cancers may allow clinicians to predict resistance to a number of commonly used therapies. Moreover, understanding the mechanism(s) of action GRP78 uses to regulate chemoresistance will accelerate the development of more efficacious treatment strategies that target GRP78 to abrogate chemoresistance. RESULTS: This mini-review highlighted the roles of targets downstream of GRP78 and explored their mechanisms related to cellular survival. Furthermore, a summary of the connection between GRP78 expression and patient prognosis was provided. Lastly, strategies for targeting GRP78, in order to improve treatment efficacy, was explored. CONCLUSIONS: GRP78 maintains an important role in regulating signaling pathways that control cell survival and this review draws attention to its value as a prognostic marker and therapeutic target.


Subject(s)
Antineoplastic Agents/pharmacology , Heat-Shock Proteins/metabolism , Neoplasms/drug therapy , Animals , Cell Survival , Drug Resistance, Neoplasm , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress , Humans , Molecular Targeted Therapy , Neoplasms/pathology , Prognosis , Signal Transduction , Unfolded Protein Response
7.
Methods Mol Biol ; 1580: 59-70, 2017.
Article in English | MEDLINE | ID: mdl-28439826

ABSTRACT

MicroRNA detection and quantification are commonly explored techniques for diagnostic and prognostic predictions. Typically, microRNAs are extracted and purified from a biological source, converted into complementary DNA (cDNA), and amplified using real time polymerase chain reaction (RT-PCR). The number of RT-PCR cycles required to reach the threshold of detection provides a relative quantification of the target microRNA when this data is normalized to the quantity of a control microRNA. This methodology has several drawbacks, including the need to artificially amplify the target microRNA for detection as well as quantification errors that can occur due to expression level differences of the control microRNAs for normalization in various sample sources. Here, we provide a technique to quantify actual concentrations of target microRNAs directly from any biological source without the requirement of these additional steps. In addition, we describe an alternative approach for obtaining exosomal microRNAs directly from biological samples without the use of harsh detergents and RNA isolation.


Subject(s)
Biosensing Techniques/methods , Exosomes/chemistry , Membranes, Artificial , MicroRNAs/analysis , Microfluidic Analytical Techniques/methods , Sound , Animals , Biosensing Techniques/instrumentation , Equipment Design , Exosomes/genetics , Humans , Ion Exchange , MicroRNAs/genetics , MicroRNAs/isolation & purification , Microfluidic Analytical Techniques/instrumentation
8.
Oncotarget ; 8(10): 16473-16487, 2017 Mar 07.
Article in English | MEDLINE | ID: mdl-28146435

ABSTRACT

Pancreatic ductal adenocarcinoma (PDAC) has a 5-year survival rate of 7%. This dismal prognosis is largely due to the inability to diagnose the disease before metastasis occurs. Tumor cell dissemination occurs early in PDAC. While it is known that inflammation facilitates this process, the underlying mechanisms responsible for this progression have not been fully characterized. Here, we functionally test the role of metastasis suppressor 1 (MTSS1) in PDAC. Despite evidence showing that MTSS1 could be important for regulating metastasis in many different cancers, its function in PDAC has not been studied. Here, we show that loss of MTSS1 leads to increased invasion and migration in PDAC cell lines. Moreover, PDAC cells treated with cancer-associated fibroblast-conditioned media also have increased metastatic potential, which is augmented by loss of MTSS1. Finally, overexpression of MTSS1 in PDAC cell lines leads to a loss of migratory potential in vitro and an increase in overall survival in vivo. Collectively, our data provide insight into an important role for MTSS1 in suppressing tumor cell invasion and migration driven by the tumor microenvironment and suggest that therapeutic strategies aimed at increasing MTSS1 levels may effectively slow the development of metastatic lesions, increasing survival of patients with PDAC.


Subject(s)
Carcinoma, Pancreatic Ductal/pathology , Microfilament Proteins/deficiency , Neoplasm Proteins/deficiency , Pancreatic Neoplasms/pathology , Animals , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Cell Line, Tumor , Gene Knockdown Techniques , HEK293 Cells , Heterografts , Humans , Male , Mice , Mice, Inbred NOD , Mice, SCID , Microfilament Proteins/genetics , Microfilament Proteins/metabolism , Neoplasm Invasiveness , Neoplasm Metastasis , Neoplasm Proteins/genetics , Neoplasm Proteins/metabolism , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/metabolism , Transfection , Tumor Microenvironment
9.
Mol Cancer Ther ; 15(5): 1043-52, 2016 05.
Article in English | MEDLINE | ID: mdl-26939701

ABSTRACT

The prognosis for patients with pancreatic ductal adenocarcinoma (PDAC) is dismal. Although gemcitabine (GEM) is the standard chemotherapeutic agent for adjuvant therapy of resectable PDAC, recurrent disease is observed in an alarming number of GEM-treated patients. Regardless of the adjuvant therapy, the vast majority of patients treated with chemotherapy after surgical resection show tumor recurrence. A better understanding of the molecular mechanisms that contribute to chemoresistance would aid the development of more effective treatment strategies. GRP78 is an endoplasmic reticulum (ER) chaperone protein that primarily resides in the lumen of the ER and is the master regulator of the unfolded protein response (UPR). Here, we report that expression of GRP78 is significantly higher in GEM-resistant PDAC compared to GEM-sensitive PDAC patient samples. We show that GRP78 induces chemoresistance in PDAC cells. Our results also show that knockdown of GRP78 reduces chemoresistance in PDAC. Finally, we found that IT-139, a ruthenium-based anticancer drug, can overcome GRP78-mediated chemoresistance. In vitro, IT-139 restores sensitivity to cytotoxic drugs in drug-resistant PDAC cells and induces twice as much cell death in combination treatment compared with GEM alone. In vivo, a single weekly IT-139 treatment in combination with GEM caused a 35% increase in median survival and a 25% increase in overall survival compared to GEM alone. Collectively, our data show that GRP78 expression promotes chemoresistance in PDAC and therapeutic strategies, blocking the activity of GRP78 increases the efficacy of currently available therapies. Mol Cancer Ther; 15(5); 1043-52. ©2016 AACR.


Subject(s)
Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/metabolism , Drug Resistance, Neoplasm/genetics , Heat-Shock Proteins/genetics , Unfolded Protein Response , Adenocarcinoma/drug therapy , Adenocarcinoma/pathology , Adult , Aged , Aged, 80 and over , Animals , Antineoplastic Agents/pharmacology , Antineoplastic Agents/therapeutic use , Carcinoma, Pancreatic Ductal/drug therapy , Carcinoma, Pancreatic Ductal/pathology , Cell Line, Tumor , Deoxycytidine/analogs & derivatives , Deoxycytidine/pharmacology , Disease Models, Animal , Drug Synergism , Endoplasmic Reticulum Chaperone BiP , Endoplasmic Reticulum Stress/genetics , Female , Gene Expression , Gene Expression Profiling , Gene Expression Regulation, Neoplastic/drug effects , Gene Knockdown Techniques , Heat-Shock Proteins/metabolism , Humans , Immunohistochemistry , Male , Middle Aged , Proto-Oncogene Proteins c-akt/genetics , Proto-Oncogene Proteins c-akt/metabolism , Survival Analysis , Xenograft Model Antitumor Assays , Gemcitabine
10.
Lab Chip ; 15(7): 1656-66, 2015 Apr 07.
Article in English | MEDLINE | ID: mdl-25690152

ABSTRACT

There has been increasing evidence that micro and messenger RNA derived from exosomes play important roles in pancreatic and other cancers. In this work, a microfluidics-based approach to the analysis of exosomal RNA is presented based on surface acoustic wave (SAW) exosome lysis and ion-exchange nanomembrane RNA sensing performed in conjunction on two separate chips. Using microRNA hsa-miR-550 as a model target and raw cell media from pancreatic cancer cell lines as a biological sample, SAW-based exosome lysis is shown to have a lysis rate of 38%, and an ion-exchange nanomembrane sensor is shown to have a limit of detection of 2 pM, with two decades of linear dynamic range. A universal calibration curve was derived for the membrane sensor and used to detect the target at a concentration of 13 pM in a SAW-lysed sample, which translates to 14 target miRNA per exosome from the raw cell media. At a total analysis time of ~1.5 h, this approach is a significant improvement over existing methods that require two overnight steps and 13 h of processing time. The platform also requires much smaller sample volumes than existing technology (~100 µL as opposed to ~mL) and operates with minimal sample loss, a distinct advantage for studies involving mouse models or other situations where the working fluid is scarce.


Subject(s)
Exosomes/chemistry , MicroRNAs/analysis , Microfluidic Analytical Techniques/instrumentation , Nanotechnology/instrumentation , Pancreatic Neoplasms/metabolism , Animals , Cell Line, Tumor , Equipment Design , Exosomes/radiation effects , Humans , Mice , Sound
11.
Mol Cancer Ther ; 11(10): 2127-37, 2012 Oct.
Article in English | MEDLINE | ID: mdl-22784710

ABSTRACT

COX-2 is upregulated in pancreatic ductal adenocarcinomas (PDAC). However, how COX-2 promotes PDAC development is unclear. While previous studies have evaluated the efficacy of COX-2 inhibition via the use of nonsteroidal anti-inflammatory drugs (NSAID) or the COX-2 inhibitor celecoxib in PDAC models, none have addressed the cell intrinsic versus microenvironment roles of COX-2 in modulating PDAC initiation and progression. We tested the cell intrinsic role of COX-2 in PDAC progression using both loss-of-function and gain-of-function approaches. Cox-2 deletion in Pdx1+ pancreatic progenitor cells significantly delays the development of PDAC in mice with K-ras activation and Pten haploinsufficiency. Conversely, COX-2 overexpression promotes early onset and progression of PDAC in the K-ras mouse model. Loss of PTEN function is a critical factor in determining lethal PDAC onset and overall survival. Mechanistically, COX-2 overexpression increases p-AKT levels in the precursor lesions of Pdx1(+); K-ras(G12D)(/+); Pten(lox)(/+) mice in the absence of Pten LOH. In contrast, Cox-2 deletion in the same setting diminishes p-AKT levels and delays cancer progression. These data suggest an important cell intrinsic role for COX-2 in tumor initiation and progression through activation of the PI3K/AKT pathway. PDAC that is independent of intrinsic COX-2 expression eventually develops with decreased FKBP5 and increased GRP78 expression, two alternate pathways leading to AKT activation. Together, these results support a cell intrinsic role for COX-2 in PDAC development and suggest that while anti-COX-2 therapy may delay the development and progression of PDAC, mechanisms known to increase chemoresistance through AKT activation must also be overcome.


Subject(s)
Cyclooxygenase 2/metabolism , Pancreatic Neoplasms/enzymology , Pancreatic Neoplasms/pathology , Precancerous Conditions/enzymology , Precancerous Conditions/pathology , Animals , Carcinoma, Pancreatic Ductal/enzymology , Carcinoma, Pancreatic Ductal/pathology , Celecoxib , Cell Membrane/drug effects , Cell Membrane/metabolism , Disease Models, Animal , Disease Progression , Endoplasmic Reticulum Chaperone BiP , Enzyme Activation/drug effects , Feedback, Physiological/drug effects , Gene Deletion , Gene Targeting , Heat-Shock Proteins/metabolism , Homeodomain Proteins , Integrases/metabolism , Life Expectancy , Mice , PTEN Phosphohydrolase/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/metabolism , Pyrazoles/chemistry , Pyrazoles/pharmacology , Sulfonamides/chemistry , Sulfonamides/pharmacology , Survival Analysis , Tacrolimus Binding Proteins/metabolism , Trans-Activators , Up-Regulation/drug effects
12.
Clin Cancer Res ; 18(5): 1352-63, 2012 Mar 01.
Article in English | MEDLINE | ID: mdl-22261810

ABSTRACT

PURPOSE: To carry out an integrative profile of human pancreatic ductal adenocarcinoma (PDAC) to identify prognosis-significant genes and their related pathways. EXPERIMENTAL DESIGN: A concordant survival-based whole genome in silico array analysis of DNA copy number, and mRNA and miRNA expression in 25 early-stage PDAC was carried out. A novel composite score simultaneously integrated gene expression with regulatory mechanisms to identify the signature genes with the most levels of prognosis-significant evidence. The predominant signaling pathways were determined via a pathway-based approach. Independent patient cohorts (n = 148 and 42) were then used as in vitro validation of the array findings. RESULTS: The composite score identified 171 genes in which expressions were able to define two prognosis subgroups (P = 3.8e-5). Eighty-eight percent (151 of 171) of the genes were regulated by prognosis-significant miRNAs. The phosphoinositide 3-kinase/AKT pathway and SRC signaling were densely populated by prognosis-significant genes and driven by genomic amplification of SRC and miRNA regulation of p85α and CBL. On tissue microarray validation (n = 148), p85α protein expression was associated with improved survival for all patients (P = 0.02), and activated P-SRC (Y418) was associated shorter survival for patients with low-grade histology tumors (P = 0.04). Interacting P-SRC and p85α revealed that they define two distinct PDAC patient subgroups (P = 0.0066). Furthering the importance of these pathways, CBL protein expression was associated with improved survival (P = 0.03) on a separate cohort (n = 42). CONCLUSIONS: These pathways and related genes may represent putative clinical biomarkers and possible targets of individualized therapy in the distinct patient subgroups they define.


Subject(s)
Carcinoma, Pancreatic Ductal/genetics , Carcinoma, Pancreatic Ductal/mortality , Gene Expression Profiling , Pancreatic Neoplasms/genetics , Pancreatic Neoplasms/mortality , Aged , Aged, 80 and over , Cluster Analysis , DNA Copy Number Variations , Female , Genomics , Humans , Male , MicroRNAs/genetics , Middle Aged , Phosphatidylinositol 3-Kinases/metabolism , Prognosis , RNA, Messenger/genetics , Reproducibility of Results , Signal Transduction , Survival Analysis , src-Family Kinases/metabolism
13.
Cancer Res ; 70(18): 7114-24, 2010 Sep 15.
Article in English | MEDLINE | ID: mdl-20807812

ABSTRACT

KRAS mutations are found in ∼90% of human pancreatic ductal adenocarcinomas (PDAC). However, mice genetically engineered to express Kras(G12D) from its endogenous locus develop PDACs only after a prolonged latency, indicating that other genetic events or pathway alterations are necessary for PDAC progression. The PTEN-controlled phosphatidylinositol 3-kinase (PI3K)/AKT signaling axis is dysregulated in later stages of PDAC. To better elucidate the role of PTEN/PI3K/AKT signaling in Kras(G12D)-induced PDAC development, we crossed Pten conditional knockout mice (Pten(lox/lox)) to mice with conditional activation of Kras(G12D). The resulting compound heterozygous mutant mice showed significantly accelerated development of acinar-to-ductal metaplasia (ADM), malignant pancreatic intraepithelial neoplasia (mPanIN), and PDAC within a year. Moreover, all mice with Kras(G12D) activation and Pten homozygous deletion succumbed to cancer by 3 weeks of age. Our data support a dosage-dependent role for PTEN, and the resulting dysregulation of the PI3K/AKT signaling axis, in both PDAC initiation and progression, and shed additional light on the signaling mechanisms that lead to the development of ADM and subsequent mPanIN and pancreatic cancer.


Subject(s)
Carcinoma, Pancreatic Ductal/enzymology , PTEN Phosphohydrolase/deficiency , Pancreatic Neoplasms/enzymology , ras Proteins/metabolism , Animals , Carcinoma, Pancreatic Ductal/genetics , Humans , Hyaluronan Receptors/biosynthesis , Mice , Mice, Inbred BALB C , Mice, Inbred C57BL , Pancreatic Neoplasms/genetics , Phosphatidylinositol 3-Kinases/metabolism , Proto-Oncogene Proteins/genetics , Proto-Oncogene Proteins/metabolism , Proto-Oncogene Proteins c-akt/metabolism , Proto-Oncogene Proteins p21(ras)/genetics , Proto-Oncogene Proteins p21(ras)/metabolism , ras Proteins/genetics
14.
J Biol Chem ; 284(18): 11755-9, 2009 May 01.
Article in English | MEDLINE | ID: mdl-19117948

ABSTRACT

Like normal stem cells, "cancer stem cells" have the capacity for indefinite proliferation and generation of new cancerous tissues through self-renewal and differentiation. Among the major intracellular signaling pathways, WNT, SHH, and NOTCH are known to be important in regulating normal stem cell activities, and their alterations are associated with tumorigenesis. It has become clear recently that PTEN (phosphatase and tensin homologue) is also critical for stem cell maintenance and that PTEN loss can cause the development of cancer stem cells and ultimately tumorigenesis.


Subject(s)
Neoplasm Proteins/metabolism , Neoplasms/metabolism , Neoplastic Stem Cells/metabolism , PTEN Phosphohydrolase/metabolism , Signal Transduction , Stem Cells/metabolism , Animals , Cell Proliferation , Humans , Neoplasm Proteins/genetics , Neoplasms/genetics , Neoplastic Stem Cells/pathology , PTEN Phosphohydrolase/genetics , Stem Cells/pathology
15.
Cancer Res ; 68(10): 3579-83, 2008 May 15.
Article in English | MEDLINE | ID: mdl-18483238

ABSTRACT

Ionizing radiation (IR) therapy is one of the most commonly used treatments for cancer patients. The responses of tumor cells to IR are often tissue specific and depend on pathway aberrations present in the tumor. Identifying molecules and mechanisms that sensitize tumor cells to IR provides new potential therapeutic strategies for cancer treatment. In this study, we used two genetically engineered mouse carcinoma models, brain choroid plexus carcinoma (CPC) and prostate, to test the effect of inactivating gadd45a, a DNA damage response p53 target gene, on tumor responses to IR. We show that gadd45a deficiency significantly increases tumor cell death after radiation. Effect on survival was assessed in the CPC model and was extended in IR-treated mice with gadd45a deficiency compared with those expressing wild-type gadd45a. These studies show a significant effect of gadd45a inactivation in sensitizing tumor cells to IR, implicating gadd45a as a potential drug target in radiotherapy management.


Subject(s)
Cell Cycle Proteins/genetics , Cell Cycle Proteins/physiology , Gene Expression Regulation, Neoplastic , Nuclear Proteins/genetics , Nuclear Proteins/physiology , Skin Neoplasms/genetics , Animals , Brain Neoplasms/metabolism , Cell Death , Cell Proliferation , Cell Survival , Disease Models, Animal , Heterozygote , Male , Mice , Mice, Transgenic , Prostatic Neoplasms/metabolism , Radiation, Ionizing , Skin Neoplasms/metabolism , Skin Neoplasms/radiotherapy
16.
Cell ; 123(6): 1001-11, 2005 Dec 16.
Article in English | MEDLINE | ID: mdl-16360031

ABSTRACT

Our understanding of cancer has largely come from the analysis of aberrations within the tumor cell population. Yet it is increasingly clear that the tumor microenvironment can significantly influence tumorigenesis. For example, the mesenchyme can support the growth of tumorigenic epithelium. However, whether fibroblasts are subject to genetic/epigenetic changes as a result of selective pressures conferred by oncogenic stress in the epithelium has not been experimentally assessed. Recent analyses of some human carcinomas have shown tumor-suppressor gene mutations within the stroma, suggesting that the interplay among multiple cell types can select for aberrations nonautonomously during tumor progression. We demonstrate that this indeed occurs in a mouse model of prostate cancer where epithelial cell cycle disruption via cell-specific inhibition of pRb function induces a paracrine p53 response that suppresses fibroblast proliferation in associated stroma. This interaction imposes strong selective pressure yielding a highly proliferative mesenchyme that has undergone p53 loss.


Subject(s)
Epithelial Cells/pathology , Fibroblasts/pathology , Mutation/genetics , Stromal Cells/pathology , Tumor Suppressor Protein p53/genetics , Actins/analysis , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Adenocarcinoma/pathology , Animals , Antigens, Polyomavirus Transforming/genetics , Cell Proliferation , Connective Tissue/pathology , Disease Models, Animal , Epithelial Cells/metabolism , Fibroblasts/metabolism , Gene Deletion , Genotype , Keratin-8 , Keratins/analysis , Loss of Heterozygosity/genetics , Male , Mice , Mice, Knockout , Mice, Transgenic , Models, Biological , Paracrine Communication , Prostate/metabolism , Prostate/pathology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Prostatic Neoplasms/pathology , Retinoblastoma Protein/metabolism , S100 Calcium-Binding Protein A4 , S100 Proteins/analysis , Stromal Cells/metabolism , Tumor Suppressor Protein p53/metabolism
17.
Cancer Res ; 65(22): 10243-54, 2005 Nov 15.
Article in English | MEDLINE | ID: mdl-16288012

ABSTRACT

Because each change in the evolution of a cancer is predicated on the effects of previous events, a full understanding of selective changes and their effect on tumor progression can only be understood in the context of appropriate initiating events. Here, we define the effect of pRb function inactivation in prostate epithelium on both the initiation of prostate cancer and the establishment of selective pressures that lead to diminished Pten function and tumor evolution. Using genetically engineered mice, we show that inactivation of the pRb family proteins (Rb/p107/p130) induces epithelial proliferation and apoptosis and is sufficient to produce prostatic intraepithelial neoplasia (PIN) lesions. Over time, adenocarcinomas develop in all mice with no evidence of neuroendocrine tumors. Apoptosis is dependent on Pten function and not p53, unlike other epithelial cell types tested previously. Consequently, Pten hemizygosity reduces apoptosis by 50%, accelerating progression to adenocarcinomas with heterogeneous composition. Heterogeneity is associated with concurrent Pten haploinsufficiency and focal selective progression to complete Pten loss, which yields distinct tumor properties. Given that this analysis models the apparent timing of highly penetrant events in human prostate cancer, observed effects may recapitulate the natural evolution of prostate cancer development.


Subject(s)
Adenocarcinoma/pathology , Cell Transformation, Neoplastic/pathology , Prostatic Neoplasms/pathology , Retinoblastoma Protein/antagonists & inhibitors , Adenocarcinoma/genetics , Adenocarcinoma/metabolism , Animals , Apoptosis/physiology , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Models, Animal , Disease Progression , Epithelial Cells/metabolism , Epithelial Cells/pathology , Female , Male , Mice , Mice, Transgenic , PTEN Phosphohydrolase/antagonists & inhibitors , PTEN Phosphohydrolase/genetics , PTEN Phosphohydrolase/metabolism , PTEN Phosphohydrolase/physiology , Prostatic Neoplasms/genetics , Prostatic Neoplasms/metabolism , Retinoblastoma Protein/genetics , Retinoblastoma Protein/metabolism , Retinoblastoma Protein/physiology , Retinoblastoma-Like Protein p107/antagonists & inhibitors , Retinoblastoma-Like Protein p107/genetics , Retinoblastoma-Like Protein p107/metabolism , Retinoblastoma-Like Protein p107/physiology , Retinoblastoma-Like Protein p130/antagonists & inhibitors , Retinoblastoma-Like Protein p130/genetics , Retinoblastoma-Like Protein p130/metabolism , Retinoblastoma-Like Protein p130/physiology
18.
Phys Rev Lett ; 89(17): 174501, 2002 Oct 21.
Article in English | MEDLINE | ID: mdl-12398673

ABSTRACT

Length scales are determined that govern the behavior at small separations of the correlations of fluid-particle acceleration, viscous force, and pressure gradient. The length scales and an associated universal constant are quantified on the basis of published data. The length scale governing pressure spectra at high wave numbers is discussed. Fluid-particle acceleration correlation is governed by two length scales: one arises from the pressure gradient, the other from the viscous force.

SELECTION OF CITATIONS
SEARCH DETAIL
...