Your browser doesn't support javascript.
loading
Montrer: 20 | 50 | 100
Résultats 1 - 20 de 37
Filtrer
1.
Brain Sci ; 11(5)2021 Apr 30.
Article de Anglais | MEDLINE | ID: mdl-33946285

RÉSUMÉ

It is important to develop minimally invasive biomarker platforms to help in the identification and monitoring of patients with Alzheimer's disease (AD). Assisting in the understanding of biochemical mechanisms as well as identifying potential novel biomarkers and therapeutic targets would be an added benefit of such platforms. This study utilizes a simplified and novel serum profiling platform, using mass spectrometry (MS), to help distinguish AD patient groups (mild and moderate) and controls, as well as to aid in understanding of biochemical phenotypes and possible disease development. A comparison of discriminating sera mass peaks between AD patients and control individuals was performed using leave one [serum sample] out cross validation (LOOCV) combined with a novel peak classification valuation (PCV) procedure. LOOCV/PCV was able to distinguish significant sera mass peak differences between a group of mild AD patients and control individuals with a p value of 10-13. This value became non-significant (p = 0.09) when the same sera samples were randomly allocated between the two groups and reanalyzed by LOOCV/PCV. This is indicative of physiological group differences in the original true-pathology binary group comparison. Similarities and differences between AD patients and traumatic brain injury (TBI) patients were also discernable using this novel LOOCV/PCV platform. MS/MS peptide analysis was performed on serum mass peaks comparing mild AD patients with control individuals. Bioinformatics analysis suggested that cell pathways/biochemical phenotypes affected in AD include those involving neuronal cell death, vasculature, neurogenesis, and AD/dementia/amyloidosis. Inflammation, autoimmunity, autophagy, and blood-brain barrier pathways also appear to be relevant to AD. An impaired VWF/ADAMTS13 vasculature axis with connections to F8 (factor VIII) and LRP1 and NOTCH1 was indicated and is proposed to be important in AD development.

2.
PLoS One ; 15(8): e0234539, 2020.
Article de Anglais | MEDLINE | ID: mdl-32756554

RÉSUMÉ

Diabetes Mellitus (DM) accelerates coronary artery disease (CAD) and atherosclerosis, the causes of most heart attacks. The biomolecules involved in these inter-related disease processes are not well understood. This study analyzes biomolecules in the sera of patients with CAD, with and without type (T) 2DM, who are about to undergo coronary artery bypass graft (CABG) surgery. The goal is to develop methodology to help identify and monitor CAD patients with and without T2DM, in order to better understand these phenotypes and to glean relationships through analysis of serum biomolecules. Aorta, fat, muscle, and vein tissues from CAD T2DM patients display diabetic-related histologic changes (e.g., lipid accumulation, fibrosis, loss of cellularity) when compared to non-diabetic CAD patients. The patient discriminatory methodology utilized is serum biomolecule mass profiling. This mass spectrometry (MS) approach is able to distinguish the sera of a group of CAD patients from controls (p value 10-15), with the CAD group containing both T2DM and non-diabetic patients. This result indicates the T2DM phenotype does not interfere appreciably with the CAD determination versus control individuals. Sera from a group of T2DM CAD patients however are distinguishable from non-T2DM CAD patients (p value 10-8), indicating it may be possible to examine the T2DM phenotype within the CAD disease state with this MS methodology. The same serum samples used in the CAD T2DM versus non-T2DM binary group comparison were subjected to MS/MS peptide structure analysis to help identify potential biochemical and phenotypic changes associated with CAD and T2DM. Such peptide/protein identifications could lead to improved understanding of underlying mechanisms, additional biomarkers for discriminating and monitoring these disease conditions, and potential therapeutic targets. Bioinformatics/systems biology analysis of the peptide/protein changes associated with CAD and T2DM suggested cell pathways/systems affected include atherosclerosis, DM, fibrosis, lipogenesis, loss of cellularity (apoptosis), and inflammation.


Sujet(s)
Maladie des artères coronaires/sang , Maladie des artères coronaires/complications , Diabète de type 2/sang , Diabète de type 2/complications , Angiopathies diabétiques/sang , Adulte , Sujet âgé , Marqueurs biologiques/sang , Protéines du sang/métabolisme , Études cas-témoins , Pontage aortocoronarien , Maladie des artères coronaires/chirurgie , Études transversales , Angiopathies diabétiques/chirurgie , Femelle , Humains , Mâle , Adulte d'âge moyen , Phénotype , Études rétrospectives , Spectrométrie de masse ESI , Biologie des systèmes , Spectrométrie de masse en tandem
3.
J Agric Food Chem ; 67(51): 14027-14037, 2019 Dec 26.
Article de Anglais | MEDLINE | ID: mdl-31771323

RÉSUMÉ

Wheat consumption has declined amid growing concerns about gluten-sensitivity. To determine if genetic manipulation of wheat contributes to systemic and localized gut inflammation, we compared the effects of the modern variety Gallagher and a blend of two heirloom varieties, Turkey and Kharkof, on measures of gut inflammation, structural characteristics, and barrier integrity under normal and Western diet (WD) conditions in C57BL/6 mice. Indicators of gut inflammation, including lymphocyte infiltration and cytokine expression, were largely unaffected by WD or wheat, although WD elevated interferon-γ (Ifng) and heirloom varieties modestly reduced interleukin-17 (Il17) in the context of WD. WD negatively affected jejunal villi structure, while the modern variety improved villi structure in the ileum. Relative mRNA and tight junction proteins and serum lipopolysaccharide binding protein were unaltered by WD or wheat. These findings indicate that the modern variety did not compromise barrier function or contribute to gut inflammation compared to its heirloom predecessor.


Sujet(s)
Tube digestif/métabolisme , Triticum/métabolisme , Animaux , Cytokines/génétique , Cytokines/immunologie , Tube digestif/immunologie , Iléum/immunologie , Iléum/métabolisme , Interféron gamma , Interleukine-17/génétique , Interleukine-17/immunologie , Mâle , Souris , Souris de lignée C57BL , Triticum/classification
4.
Cancer Invest ; 35(9): 573-585, 2017 Oct 21.
Article de Anglais | MEDLINE | ID: mdl-28949774

RÉSUMÉ

A stage I non-small cell lung cancer (NSCLC) serum profiling platform is presented which is highly efficient and accurate. Test sensitivity (0.95) for stage I NSCLC is the highest reported so far. Test metrics are reported for discriminating stage I adenocarcinoma vs squamous cell carcinoma subtypes. Blinded analysis identified 23 out of 24 stage I NSCLC and control serum samples. Group-discriminating mass peaks were targeted for tandem mass spectrometry peptide/protein identification, and yielded a lung cancer phenotype. Bioinformatic analysis revealed a novel lymphocyte adhesion pathway involved with early-stage lung cancer.


Sujet(s)
Adénocarcinome/sang , Marqueurs biologiques tumoraux/sang , Carcinome pulmonaire non à petites cellules/sang , Carcinome épidermoïde/sang , Tumeurs du poumon/sang , Protéomique/méthodes , Spectrométrie de masse en tandem , Adénocarcinome/immunologie , Adénocarcinome/anatomopathologie , Adénocarcinome pulmonaire , Sujet âgé , Sujet âgé de 80 ans ou plus , Carcinome pulmonaire non à petites cellules/immunologie , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome épidermoïde/immunologie , Carcinome épidermoïde/anatomopathologie , Études cas-témoins , Adhérence cellulaire , Biologie informatique , Bases de données de protéines , Diagnostic différentiel , Femelle , Humains , Tumeurs du poumon/immunologie , Tumeurs du poumon/anatomopathologie , Lymphocytes/immunologie , Lymphocytes/métabolisme , Mâle , Adulte d'âge moyen , Stadification tumorale , Phénotype , Valeur prédictive des tests
5.
J Clin Invest ; 126(2): 706-20, 2016 Feb.
Article de Anglais | MEDLINE | ID: mdl-26731476

RÉSUMÉ

Histone demethylase upregulation has been observed in human cancers, yet it is unknown whether this is a bystander event or a driver of tumorigenesis. We found that overexpression of lysine-specific demethylase 4A (KDM4A, also known as JMJD2A) was positively correlated with Gleason score and metastasis in human prostate tumors. Overexpression of JMJD2A resulted in the development of prostatic intraepithelial neoplasia in mice, demonstrating that JMJD2A can initiate prostate cancer development. Moreover, combined overexpression of JMJD2A and the ETS transcription factor ETV1, a JMJD2A-binding protein, resulted in prostate carcinoma formation in mice haplodeficient for the phosphatase and tensin homolog (Pten) tumor-suppressor gene. Additionally, JMJD2A cooperated with ETV1 to increase expression of yes associated protein 1 (YAP1), a Hippo pathway component that itself was associated with prostate tumor aggressiveness. ETV1 facilitated the recruitment of JMJD2A to the YAP1 promoter, leading to changes in histone lysine methylation in a human prostate cancer cell line. Further, YAP1 expression largely rescued the growth inhibitory effects of JMJD2A depletion in prostate cancer cells, indicating that YAP1 is a downstream effector of JMJD2A. Taken together, these data reveal a JMJD2A/ETV1/YAP1 axis that promotes prostate cancer initiation and that may be a suitable target for therapeutic inhibition.


Sujet(s)
Transformation cellulaire néoplasique/métabolisme , Protéines de liaison à l'ADN/métabolisme , Histone Demethylases/métabolisme , Jumonji Domain-Containing Histone Demethylases/métabolisme , Protéines tumorales/métabolisme , Tumeurs de la prostate/métabolisme , Facteurs de transcription/métabolisme , Protéines adaptatrices de la transduction du signal/génétique , Protéines adaptatrices de la transduction du signal/métabolisme , Animaux , Protéines du cycle cellulaire , Lignée cellulaire tumorale , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/anatomopathologie , Protéines de liaison à l'ADN/génétique , Femelle , Histone Demethylases/génétique , Humains , Jumonji Domain-Containing Histone Demethylases/génétique , Mâle , Souris , Souris transgéniques , Protéines tumorales/génétique , Phosphoprotéines/génétique , Phosphoprotéines/métabolisme , Tumeurs de la prostate/génétique , Facteurs de transcription/génétique , Protéines de signalisation YAP
6.
Int J Clin Exp Med ; 9(6): 10123-10134, 2016.
Article de Anglais | MEDLINE | ID: mdl-28883898

RÉSUMÉ

PSMD10, also known as gankyrin, is associated with the proteasome and has been shown to be an oncoprotein in the liver. Here, we report that PSMD10 expression is stimulated by the histone demethylase JMJD2A/KDM4A and its interaction partner, the ETV1 transcription factor, in LNCaP prostate cancer cells. Global analysis of expression patterns revealed that PSMD10 mRNA levels are positively correlated with those of both JMJD2A and ETV1. In human prostate tumors, PSMD10 is highly overexpressed at the protein level and correlates with JMJD2A overexpression; further, PSMD10 expression is enhanced in the prostates of transgenic JMJD2A mice. Moreover, PSMD10 is particularly overexpressed in high Gleason score prostate tumors. Downregulation of PSMD10 in LNCaP prostate cancer cells impaired their growth, indicating that PSMD10 may exert a pro-oncogenic function in the prostate. Lastly, we observed that PSMD10 expression is correlated to YAP1, a component of the Hippo signaling pathway and whose gene promoter is regulated by JMJD2A, and that PSMD10 can cooperate with YAP1 in stimulating LNCaP cell growth. Altogether, these data indicate that PSMD10 is a novel downstream effector of JMJD2A and suggest that inhibition of the JMJD2A histone demethylase by small molecule drugs may be effective to curtail the oncogenic activity of PSMD10 in various PSMD10-overexpressing tumors.

7.
PLoS One ; 10(8): e0135561, 2015.
Article de Anglais | MEDLINE | ID: mdl-26270561

RÉSUMÉ

Gastrointestinal (GI) mucosal damage is a devastating adverse effect of radiation therapy. We have recently reported that expression of Dclk1, a Tuft cell and tumor stem cell (TSC) marker, 24h after high dose total-body gamma-IR (TBI) can be used as a surrogate marker for crypt survival. Dietary pectin has been demonstrated to possess chemopreventive properties, whereas its radioprotective property has not been studied. The aim of this study was to determine the effects of dietary pectin on ionizing radiation (IR)-induced intestinal stem cell (ISC) deletion, crypt and overall survival following lethal TBI. C57BL/6 mice received a 6% pectin diet and 0.5% pectin drinking water (pre-IR mice received pectin one week before TBI until death; post-IR mice received pectin after TBI until death). Animals were exposed to TBI (14 Gy) and euthanized at 24 and 84h post-IR to assess ISC deletion and crypt survival respectively. Animals were also subjected to overall survival studies following TBI. In pre-IR treatment group, we observed a three-fold increase in ISC/crypt survival, a two-fold increase in Dclk1+ stem cells, increased overall survival (median 10d vs. 7d), and increased expression of Dclk1, Msi1, Lgr5, Bmi1, and Notch1 (in small intestine) post-TBI in pectin treated mice compared to controls. We also observed increased survival of mice treated with pectin (post-IR) compared to controls. Dietary pectin is a radioprotective agent; prevents IR-induced deletion of potential reserve ISCs; facilitates crypt regeneration; and ultimately promotes overall survival. Given the anti-cancer activity of pectin, our data support a potential role for dietary pectin as an agent that can be administered to patients receiving radiation therapy to protect against radiation-induces mucositis.


Sujet(s)
Inflammation muqueuse/prévention et contrôle , Pectine/administration et posologie , Lésions radiques expérimentales/prévention et contrôle , Radioprotecteurs/administration et posologie , Cellules souches/effets des médicaments et des substances chimiques , Animaux , Survie cellulaire/effets des médicaments et des substances chimiques , Compléments alimentaires/analyse , Kinases de type doublecortine , Femelle , Muqueuse gastrique/cytologie , Muqueuse gastrique/métabolisme , Régulation de l'expression des gènes/effets des médicaments et des substances chimiques , Souris , Souris de lignée C57BL , Inflammation muqueuse/diétothérapie , Inflammation muqueuse/étiologie , Inflammation muqueuse/anatomopathologie , Pectine/pharmacologie , Protein-Serine-Threonine Kinases/génétique , Protein-Serine-Threonine Kinases/métabolisme , Lésions radiques expérimentales/diétothérapie , Lésions radiques expérimentales/anatomopathologie , Radioprotecteurs/pharmacologie , Cellules souches/métabolisme , Cellules souches/effets des radiations , Analyse de survie , Irradiation corporelle totale
8.
J Nutr Metab ; 2015: 758080, 2015.
Article de Anglais | MEDLINE | ID: mdl-26146567

RÉSUMÉ

Type 2 diabetes mellitus (T2DM) represents a complex clinical scenario of altered energy metabolism and increased fracture incidence. The C57BL/6 mouse model of diet-induced obesity has been used to study the mechanisms by which altered glucose homeostasis affects bone mass and quality, but genetic variations in substrains of C57BL/6 may have confounded data interpretation. This study investigated the long-term metabolic and skeletal consequences of two commonly used C57BL/6 substrains to a high fat (HF) diet. Male C57BL/6J, C57BL/6N, and the negative control strain, C3H/HeJ, mice were fed a control or HF diet for 24 wks. C57BL/6N mice on a HF diet demonstrated an increase in plasma insulin and blood glucose as early as 4 wk, whereas these responses were delayed in the C57BL/6J mice. The C57BL/6N mice exhibited more severe hepatic steatosis and inflammation. Only the C57BL/6N mice lost significant trabecular bone in response to the high fat diet. The C3H/HeJ mice were protected from bone loss. The data show that C57BL/6J and C57BL/6N mice differ in their metabolic and skeletal response when fed a HF diet. These substrain differences should be considered when designing experiments and are likely to have implications on data interpretation and reproducibility.

9.
Cancer Lett ; 359(2): 314-24, 2015 Apr 10.
Article de Anglais | MEDLINE | ID: mdl-25637792

RÉSUMÉ

Blood tests are needed to aid in the early detection of pancreatic ductal adenocarcinoma (PDAC), and monitoring pancreatitis development into malignancy especially in high risk patients. This study exhibits efforts and progress toward developing such blood tests, using electrospray-mass spectrometry (MS) serum profiling to distinguish patients with early-stage PDAC or pancreatitis from each other and from controls. Identification of significant serum mass peak differences between these individuals was performed using t tests and "leave one out" cross validation. Serum mass peak distributions of control individuals were distinguished from those of patients with chronic pancreatitis or early-stage PDAC with P values <10(-15), and patients with chronic pancreatitis were distinguished from those of patients with early-stage PDAC with a P value <10(-12). Sera from 12 out of 12 patients with PDAC stages I, IIA and IIB were blindly validated from controls. Tandem MS/MS identified a cancer phenotype with elements of PDAC involved in early-stage PDAC/control discrimination. These studies indicate electrospray-MS mass profiling can detect serum changes in patients with pancreatitis or early-stage pancreatic cancer. Such technology has the potential to aid in early detection of pancreatic cancer, biomarker development, and in monitoring development of pancreatitis into PDAC.


Sujet(s)
Marqueurs biologiques tumoraux/sang , Carcinome du canal pancréatique/diagnostic , Tumeurs du pancréas/diagnostic , Pancréatite chronique/diagnostic , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Carcinome du canal pancréatique/sang , Diagnostic différentiel , Dépistage précoce du cancer , Femelle , Humains , Mâle , Adulte d'âge moyen , Pancréas/métabolisme , Tumeurs du pancréas/sang , Pancréatite chronique/sang , Spectrométrie de masse ESI , Spectrométrie de masse en tandem
10.
Dig Dis Sci ; 60(2): 509-13, 2015 Feb.
Article de Anglais | MEDLINE | ID: mdl-25283374

RÉSUMÉ

BACKGROUND: Doublecortin-like kinase 1 (DCLK1), a putative tumor stem cell marker has been shown to be highly expressed in the stromal and epithelial compartments in colon and pancreatic cancer as well as Barrett's esophagus (BE) and esophageal adenocarcinoma (EAC). AIM: To prospectively investigate whether the immunohistochemical expression of DCLK1 was associated with detectable DCLK1 plasma expression in patients with existing BE and EAC. METHODS: Immunohistochemistry was performed on paraffin-embedded sections using DCLK1 antibody and scored based on staining intensity and tissue involvement. Purified human plasma samples were subjected to Western blot and ELISA analysis. RESULTS: Forty (40) patients were enrolled: 10 controls (normal endoscopy) and 30 with BE/EAC (13 nondysplastic BE [NDBE], 9 dysplastic BE [DBE] and 8 EAC). Mean epithelial DCLK1 staining was as follows: controls = 0.11, NDBE = 3.83, DBE = 6.0, EAC = 7.17. Mean stromal DCLK1 staining was as follows: NDBE = 5.83, DBE = 5.375, EAC = 10.83. DCLK1 was detected by plasma Western blot in 1 control and in all patients with BE/EAC p < 0.0005. Plasma DCLK1 was elevated by ELISA in EAC compared to other groups, p < 0.05. CONCLUSIONS: Increased expression of DCLK1 was observed in the epithelium, stroma and plasma of patients with BE/EAC. Furthermore, the presence of detectable DCLK1 in plasma of BE/EAC patients may provide a less invasive, detection tool in those patients as well as represent a novel molecular marker distinguishing between normal esophageal mucosa and BE or EAC.


Sujet(s)
Adénocarcinome/enzymologie , Oesophage de Barrett/enzymologie , Marqueurs biologiques tumoraux/sang , Tumeurs de l'oesophage/enzymologie , Protéines et peptides de signalisation intracellulaire/sang , Protein-Serine-Threonine Kinases/sang , Adénocarcinome/sang , Adénocarcinome/anatomopathologie , Oesophage de Barrett/sang , Oesophage de Barrett/anatomopathologie , Technique de Western , Études cas-témoins , Kinases de type doublecortine , Test ELISA , Cellules épithéliales/enzymologie , Tumeurs de l'oesophage/sang , Tumeurs de l'oesophage/anatomopathologie , Oesophagoscopie , Humains , Immunohistochimie , Valeur prédictive des tests , Pronostic , Études prospectives , Cellules stromales/enzymologie , Régulation positive
11.
Oncotarget ; 5(19): 9269-80, 2014 Oct 15.
Article de Anglais | MEDLINE | ID: mdl-25211188

RÉSUMÉ

Doublecortin-like kinase 1 (Dclk1) is overexpressed in many cancers including colorectal cancer (CRC) andit specifically marks intestinal tumor stem cells. However, the role of Dclk1 in intestinal tumorigenesis in Apc mutant conditions is still poorly understood. We demonstrate that Dclk1 expression and Dclk1+ cells are significantly increased in the intestinal epithelium of elderly ApcMin/+ mice compared to young ApcMin/+ mice and wild type mice. Intestinal epithelial cells of ApcMin/+ mice demonstrate increased pluripotency, self-renewing ability, and EMT. Furthermore, miRNAs are dysregulated, expression of onco-miRNAs are significantly increased with decreased tumor suppressor miRNAs. In support of these findings, knockdown of Dclk1 in elderly ApcMin/+ mice attenuates intestinal adenomas and adenocarcinoma by decreasing pluripotency, EMT and onco-miRNAs indicating that Dclk1 overexpression facilitates intestinal tumorigenesis. Knocking down Dclk1 weakens Dclk1-dependent intestinal processes for tumorigenesis. This study demonstrates that Dclk1 is critically involved in facilitating intestinal tumorigenesis by enhancing pluripotency and EMT factors in Apc mutant intestinal tumors and it also provides a potential therapeutic target for the treatment of colorectal cancer.


Sujet(s)
Protéine de la polypose adénomateuse colique/génétique , Transition épithélio-mésenchymateuse/génétique , Tumeurs de l'intestin/anatomopathologie , Cellules souches tumorales/anatomopathologie , Protein-Serine-Threonine Kinases/génétique , Adénocarcinome/génétique , Adénomes/génétique , Animaux , Transformation cellulaire néoplasique/génétique , Kinases de type doublecortine , Cellules épithéliales/anatomopathologie , Muqueuse intestinale/métabolisme , Tumeurs de l'intestin/génétique , Intestins/cytologie , Souris , Souris de lignée C57BL , Souris transgéniques , microARN/génétique , Protein-Serine-Threonine Kinases/biosynthèse , Interférence par ARN , Petit ARN interférent , Sphéroïdes de cellules , Cellules cancéreuses en culture
12.
Cancer Invest ; 32(4): 136-43, 2014 May.
Article de Anglais | MEDLINE | ID: mdl-24579933

RÉSUMÉ

Serum mass profiling can discern physiological changes associated with specific disease states and their progression. Sera (86 total) from control individuals and patients with stage I nonsmall cell lung cancer or benign small pulmonary nodules were discriminated retrospectively by serum changes discerned by mass profiling. Control individuals were distinguished from patients with Stage I lung cancer or benign nodules with test sensitivities of 89% and 83%. Lung cancer patients versus those with benign nodules were distinguished with 80% sensitivity. This study exhibits progress toward a minimally-invasive aid in early detection of lung cancer and monitoring small pulmonary nodules for malignancy.


Sujet(s)
Marqueurs biologiques tumoraux/sang , Carcinome pulmonaire non à petites cellules/diagnostic , Tumeurs du poumon/diagnostic , Nodules pulmonaires multiples/diagnostic , Protéomique , Nodule pulmonaire solitaire/diagnostic , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Biopsie , Carcinome pulmonaire non à petites cellules/sang , Carcinome pulmonaire non à petites cellules/anatomopathologie , Diagnostic différentiel , Dépistage précoce du cancer , Femelle , Humains , Tumeurs du poumon/sang , Tumeurs du poumon/anatomopathologie , Mâle , Adulte d'âge moyen , Nodules pulmonaires multiples/sang , Nodules pulmonaires multiples/anatomopathologie , Stadification tumorale , Valeur prédictive des tests , Protéomique/méthodes , Études rétrospectives , Nodule pulmonaire solitaire/sang , Nodule pulmonaire solitaire/anatomopathologie , Spectrométrie de masse ESI , Tomodensitométrie , Charge tumorale
13.
PLoS One ; 8(9): e73940, 2013.
Article de Anglais | MEDLINE | ID: mdl-24040120

RÉSUMÉ

Stem cell pluripotency, angiogenesis and epithelial-mesenchymal transition (EMT) have been shown to be significantly upregulated in pancreatic ductal adenocarcinoma (PDAC) and many other aggressive cancers. The dysregulation of these processes is believed to play key roles in tumor initiation, progression, and metastasis, and is contributory to PDAC being the fourth leading cause of cancer-related deaths in the US. The tumor suppressor miRNA miR-145 downregulates critical pluripotency factors and oncogenes and results in repressed metastatic potential in PDAC. Additionally, the miR-200 family regulates several angiogenic factors which have been linked to metastasis in many solid tumors. We have previously demonstrated that downregulation of DCLK1 can upregulate critical miRNAs in both in vitro and in vivo cancer models and results in downregulation of c-MYC, KRAS, NOTCH1 and EMT-related transcription factors. A recent report has also shown that Dclk1 can distinguish between normal and tumor stem cells in Apc (min/+) mice and that ablation of Dclk1(+) cells resulted in regression of intestinal polyps without affecting homeostasis. Here we demonstrate that the knockdown of DCLK1 using poly(lactide-co-glycolide)-encapsulated-DCLK1-siRNA results in AsPC1 tumor growth arrest. Examination of xenograft tumors revealed, (a) increased miR-145 which results in decreased pluripotency maintenance factors OCT4, SOX2, NANOG, KLF4 as well as KRAS and RREB1; (b) increased let-7a which results in decreased pluripotency factor LIN28B; and (c) increased miR-200 which results in decreased VEGFR1, VEGFR2 and EMT-related transcription factors ZEB1, ZEB2, SNAIL and SLUG. Specificity of DCLK1 post-transcriptional regulation of the downstream targets of miR-145, miR-200 and let-7a was accomplished utilizing a luciferase-based reporter assay. We conclude that DCLK1 plays a significant master regulatory role in pancreatic tumorigenesis through the regulation of multiple tumor suppressor miRNAs and their downstream pro-tumorigenic pathways. This novel concept of targeting DCLK1 alone has several advantages over targeting single pathway or miRNA-based therapies for PDAC.


Sujet(s)
Protéines et peptides de signalisation intracellulaire/génétique , microARN/génétique , microARN/métabolisme , Néovascularisation pathologique/génétique , Tumeurs du pancréas/génétique , Tumeurs du pancréas/métabolisme , Protein-Serine-Threonine Kinases/génétique , Animaux , Lignée cellulaire tumorale , Protéines de liaison à l'ADN/génétique , Protéines de liaison à l'ADN/métabolisme , Modèles animaux de maladie humaine , Kinases de type doublecortine , Transition épithélio-mésenchymateuse/génétique , Régulation de l'expression des gènes tumoraux , Extinction de l'expression des gènes , Humains , Protéines et peptides de signalisation intracellulaire/métabolisme , Facteur-4 de type Kruppel , Souris , Cellules souches tumorales/métabolisme , Tumeurs du pancréas/anatomopathologie , Protein-Serine-Threonine Kinases/métabolisme , Maturation post-transcriptionnelle des ARN , Protéines de liaison à l'ARN , Charge tumorale/génétique , Tests d'activité antitumorale sur modèle de xénogreffe
14.
Cancer Res ; 73(16): 5110-9, 2013 Aug 15.
Article de Anglais | MEDLINE | ID: mdl-23774214

RÉSUMÉ

Overexpression of the ETS-related transcription factor ETV1 can initiate neoplastic transformation of the prostate. ETV1 activity is highly regulated by phosphorylation, but the underlying mechanisms are unknown. Here we report that all 14-3-3 proteins, with the exception of the tumor suppressor 14-3-3σ, can bind to ETV1 in a condition manner dictated by its prominent phosphorylation site S216. Non-σ 14-3-3 proteins synergized with ETV1 to activate transcription of its target genes MMP-1 and MMP-7, which regulate extracellular matrix in the prostate tumor microenvironment. S216 mutation or 14-3-3τ downregulation was sufficient to reduce ETV1 protein levels in prostate cancer cells, indicating that non-σ 14-3-3 proteins protect ETV1 from degradation. Notably, S216 mutation also decreased ETV1-dependent migration and invasion in benign prostate cells. Downregulation of 14-3-3τ reduced prostate cancer cell invasion and growth in the same manner as ETV1 attenuation. Finally, we showed that 14-3-3τ and 14-3-3ε were overexpressed in human prostate tumors. Taken together, our results showed that non-σ 14-3-3 proteins are important modulators of ETV1 function that promote prostate tumorigenesis.


Sujet(s)
Protéines 14-3-3/génétique , Marqueurs biologiques tumoraux/génétique , Protéines de liaison à l'ADN/génétique , Exoribonucleases/génétique , Tumeurs de la prostate/génétique , Protéines proto-oncogènes c-ets/génétique , Facteurs de transcription/génétique , Protéines 14-3-3/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Lignée cellulaire , Mouvement cellulaire/physiologie , Protéines de liaison à l'ADN/métabolisme , Régulation négative , Exoribonucleases/métabolisme , Régulation de l'expression des gènes tumoraux , Cellules HEK293 , Humains , Mâle , Matrix metalloproteinase 1/génétique , Matrix metalloproteinase 1/métabolisme , Matrix metalloproteinase 7/génétique , Matrix metalloproteinase 7/métabolisme , Mutation , Invasion tumorale , Phosphorylation , Tumeurs de la prostate/métabolisme , Tumeurs de la prostate/anatomopathologie , Protéines proto-oncogènes c-ets/métabolisme , Facteurs de transcription/métabolisme , Microenvironnement tumoral/génétique
15.
Int J Cancer ; 133(11): 2662-71, 2013 Dec 01.
Article de Anglais | MEDLINE | ID: mdl-23712558

RÉSUMÉ

Mass spectrometry (MS) has the unique ability to profile, in an easily accessible body tissue (peripheral blood/serum,) the sizes and relative amounts of a wide variety of biomolecules in a single platform setting. Using electrospray ionization (ESI)-MS, we distinguished individual serum from wild-type control mice from serum of mice containing an oncogenic Kras mutation, which leads to development of pancreatic ductal adenocarcinoma (PDAC) similar to that observed in humans. Identification of differences in significant ESI-MS sera mass peaks between Kras-activated mice and control mice was performed using t tests and a "nested leave one out" cross-validation procedure. Peak distributions in serum of control mice from mice with Kras-mutant-dependent PDAC were distinguished from those of pancreatic intraepithelial neoplasia (PanIN) lesions (p = 0.00024). In addition, Kras mutant mice with PDAC were distinguished from Kras mutant mice with PanIN alone (p = 0.0057). Test specificity, a measure of the false positives, was greater for the control vs. Kras mutated mice, and the test sensitivity, a measure of false negatives, was greater for the PDAC vs. PanIN containing mice. Receiver-operating characteristic (ROC) curve discriminatory values were 0.85 for both comparisons. These studies indicate ESI-MS serum mass profiling can detect physiological changes associated with pancreatic cancer initiation and development in a GEM (genetic engineered mouse) model that mimics pancreatic cancer development in humans. Such technology has the potential to aid in early detection of pancreatic cancer and in developing therapeutic drug interventions.


Sujet(s)
Adénocarcinome/génétique , Tumeurs du pancréas/sang , Protéines proto-oncogènes p21(ras)/génétique , Sérum , Adénocarcinome/sang , Animaux , Humains , Souris , Souris transgéniques/sang , Mutation , Tumeurs expérimentales/sang , Tumeurs du pancréas/génétique , Tumeurs du pancréas/anatomopathologie , Spectrométrie de masse ESI
16.
Int J Oncol ; 41(5): 1701-6, 2012 Nov.
Article de Anglais | MEDLINE | ID: mdl-22948256

RÉSUMÉ

Estrogen receptor α (ERα) plays a pivotal role in the genesis of the majority of breast tumors. Consequently, endocrine therapy is now routinely utilized in the clinic for the treatment of ERα-positive breast cancer patients. However, how ERα activity becomes dysregulated in breast cancer cells remains to be elucidated. The aim of this study was to show that the histone demethylase JMJD2A, also known as KDM4A, is capable of forming a complex with ERα in vivo. Moreover, wild-type JMJD2A, but not a catalytically impaired mutant, was able to strongly coactivate ERα-mediated transcription. Consistently, the downregulation of JMJD2A in human T47D breast cancer cells led to a decreased expression of cyclin D1, a prominent ERα target gene and cell cycle regulator. The downregulation of JMJD2A induced a reduction in the growth of T47D cells. In addition, we found that JMJD2A is overexpressed in human breast tumors both at the mRNA and protein level. Taken together, these data indicate that the overexpression of JMJD2A may contribute to breast tumor formation by stimulating ERα activity and that JMJD2A may be a breast-relevant oncoprotein. As such, small molecule drugs targeting the catalytic center of JMJD2A might be useful in breast cancer adjuvant therapy.


Sujet(s)
Tumeurs du sein/génétique , Tumeurs du sein/métabolisme , Transformation cellulaire néoplasique/génétique , Transformation cellulaire néoplasique/métabolisme , Jumonji Domain-Containing Histone Demethylases/génétique , Jumonji Domain-Containing Histone Demethylases/métabolisme , Lignée cellulaire tumorale , Prolifération cellulaire , Récepteur alpha des oestrogènes/métabolisme , Femelle , Expression des gènes , Régulation de l'expression des gènes tumoraux , Humains , Liaison aux protéines , Transcription génétique
17.
Cancer Invest ; 30(2): 189-97, 2012 Feb.
Article de Anglais | MEDLINE | ID: mdl-22149058

RÉSUMÉ

Sera mass spectrometry (MS) peak differences were analyzed from 35 ovarian cancer patients and 16 disease-free individuals. "Leave one out" cross validation was used to assign "% cancer peaks" in control and ovarian cancer sera samples. Sera MS discriminated stage I/II and stage III/V ovarian cancer patients versus controls with ROC curve area values of 0.82 and 0.92. Test sensitivities for ovarian cancer stage I/II and III/V were 80% and 93% respectively. These results indicate that MS is useful for distinguishing sera from early-stage ovarian cancer patients, and has potential as a test for early detection of this disease.


Sujet(s)
Marqueurs biologiques tumoraux/sang , Tumeurs de l'ovaire/sang , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Études cas-témoins , Survie sans rechute , Femelle , Humains , Spectrométrie de masse , Adulte d'âge moyen , Stadification tumorale , Tumeurs de l'ovaire/diagnostic , Tumeurs de l'ovaire/anatomopathologie
18.
Cancer Invest ; 30(2): 180-8, 2012 Feb.
Article de Anglais | MEDLINE | ID: mdl-22149138

RÉSUMÉ

Goals of this study were to analyze the ability of mass spectrometry serum profiling to distinguish non-small cell lung adenocarcinoma from squamous cell carcinoma patients and healthy controls. Sera were obtained from 19 adenocarcinoma patients, 24 squamous cell carcinoma patients, and 21 controls. Identifications of significant mass-to-charge ratio (m/z) peak differences between these groups were performed using t-tests. A "leave one out" cross-validation procedure yielded discriminatory lung adenocarcinoma versus squamous cell carcinoma p and ROC curve values of <.0001 and 0.92, respectively. Test sensitivity and specificity were 84% and 79%, respectively. This approach could aid in lung cancer diagnosis and sub-typing.


Sujet(s)
Adénocarcinome/sang , Carcinome épidermoïde/sang , Tumeurs du poumon/sang , Adénocarcinome/diagnostic , Adénocarcinome/anatomopathologie , Adénocarcinome pulmonaire , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Carcinome épidermoïde/diagnostic , Carcinome épidermoïde/anatomopathologie , Études cas-témoins , Diagnostic différentiel , Femelle , Humains , Tumeurs du poumon/diagnostic , Tumeurs du poumon/anatomopathologie , Mâle , Spectrométrie de masse/méthodes , Adulte d'âge moyen
19.
J Gastroenterol Hepatol ; 27(4): 773-80, 2012 Apr.
Article de Anglais | MEDLINE | ID: mdl-21916995

RÉSUMÉ

BACKGROUND AND AIM: In Barrett's esophagus (BE), the normal esophageal squamous epithelium is replaced with a specialized metaplastic columnar epithelium. BE is a premalignant lesion that can progress to esophageal adenocarcinoma (EAC). Currently, there are no early molecular indicators that would predict progression from BE to EAC. As the only permanent residents of the epithelium, stem cells have been implicated in this metaplastic progression. The aim of the present study was to determine the expression of doublecortin and CaM kinase-like-1 (DCAMKL-1) and other putative gastrointestinal stem cell markers in normal esophageal mucosa (NEM), BE, and EAC. METHODS: Human NEM, BE, EAC, and multitissue microarrays were analyzed for DCAMKL-1, and immunohistochemically scored based on staining intensity and tissue involvement, with epithelia and stroma scored separately. Total RNA isolated from BE and paired NEM was subjected to real-time reverse-transcription-polymerase chain reaction analysis for DCAMKL-1, leucine-rich repeat-containing G-protein-coupled receptor (LGR5), and Musashi-1 (Msi-1) mRNA expression. RESULTS: DCAMKL-1 was minimally expressed in squamous NEM, but increased in BE (with and without dysplasia) and EAC tissues. In EAC, we found increased stromal DCAMKL-1 staining compared to adjacent epithelia. Within the submucosa of dysplastic BE tissues, an increase in the endothelial cell expression of DCAMKL-1 was observed. Finally, an upregulation of DCAMKL-1, LGR5, and Msi-1 mRNA was seen in BE compared to squamous NEM. CONCLUSIONS: In the present study, we report the progressive increase of DCAMKL-1 expression in BE from dysplasia to EAC. Furthermore, there was an increase in putative stem cell markers DCAMKL-1, LGR5, and Msi-1 mRNA. Taken together, these data suggest that the regulation of resident stem cells might play an important role in the progression of BE to EAC.


Sujet(s)
Adénocarcinome/métabolisme , Oesophage de Barrett/métabolisme , Marqueurs biologiques tumoraux/métabolisme , Tumeurs de l'oesophage/métabolisme , Oesophage/métabolisme , Protéines et peptides de signalisation intracellulaire/métabolisme , Protein-Serine-Threonine Kinases/métabolisme , Cellules souches/métabolisme , Oesophage de Barrett/anatomopathologie , Kinases de type doublecortine , Humains , Analyse sur microréseau , Protéines de tissu nerveux/métabolisme , ARN messager/métabolisme , Protéines de liaison à l'ARN/métabolisme , Récepteurs couplés aux protéines G/métabolisme , RT-PCR , Régulation positive
20.
Lung Cancer ; 74(2): 206-11, 2011 Nov.
Article de Anglais | MEDLINE | ID: mdl-21529985

RÉSUMÉ

The goal of this study was to evaluate the usefulness of electrospray ionization-mass spectrometry (ESI-MS) technology to distinguish sera of early-stage lung cancer patients from control individuals. ESI-MS m/z (mass divided by charge) data were generated from sera of 43 non-small cell lung cancer patients (pathological stages I and II) and 21 control individuals. Identifications of m/z peak area significances between cancer and control ESI-MS sera spectra were performed using t-tests. A "leave one out" cross validation procedure, which mimics blinded sera analysis and corrects for "over-fitting" of data, yielded discriminatory cancer versus control distribution p value and ROC curve area value of <0.001 and 0.87, respectively. Analysis without the "leave one out" cross validation procedure yielded a ROC curve area of 0.99 for discrimination of sera from lung cancer patients versus control individuals. Predictive value measurements revealed overall test efficiency and sensitivity for distinguishing sera from lung cancer patients from controls (using "leave one out" cross validation) of 80% and 84%, respectively. ESI-MS serum analysis between control individuals and lung cancer patients who smoked or did not smoke had p values in ranges indicating that smoking effects are not pronounced in our analysis. These studies indicate that ESI-MS analyses of sera from early stage non-small cell lung cancer patients were helpful in distinguishing these patients from control individuals.


Sujet(s)
Carcinome pulmonaire non à petites cellules/diagnostic , Tumeurs du poumon/diagnostic , Spectrométrie de masse ESI , Adulte , Sujet âgé , Sujet âgé de 80 ans ou plus , Marqueurs biologiques tumoraux/sang , Carcinome pulmonaire non à petites cellules/anatomopathologie , Carcinome pulmonaire non à petites cellules/physiopathologie , Diagnostic différentiel , Dépistage précoce du cancer , Études de faisabilité , Femelle , Humains , Tumeurs du poumon/anatomopathologie , Tumeurs du poumon/physiopathologie , Mâle , Adulte d'âge moyen , Stadification tumorale , Valeur prédictive des tests , Courbe ROC , Sensibilité et spécificité
SÉLECTION CITATIONS
DÉTAIL DE RECHERCHE