Your browser doesn't support javascript.
loading
Show: 20 | 50 | 100
Results 1 - 20 de 37
Filter
Add more filters

Publication year range
1.
Am J Pathol ; 185(11): 2907-22, 2015 Nov.
Article in English | MEDLINE | ID: mdl-26429739

ABSTRACT

Cripto-1, a member of the epidermal growth factor-Cripto-1/FRL-1/Cryptic family, is critical for early embryonic development. Together with its ligand Nodal, Cripto-1 has been found to be associated with the undifferentiated status of mouse and human embryonic stem cells. Several studies have clearly shown that Cripto-1 is involved in regulating branching morphogenesis and epithelial-mesenchymal transition of the mammary gland both in vitro and in vivo and together with the cofactor GRP78 is critical for the maintenance of mammary stem cells ex vivo. Our previous studies showed that mammary-specific overexpression of human Cripto-1 exhibited dramatic morphological alterations in nulliparous mice mammary glands. The present study shows a novel mechanism for Cripto-1 regulation of mammary gland development through direct effects on progesterone receptor expression and pathways regulated by progesterone in the mammary gland. We demonstrate a strict temporal regulation of mouse Cripto-1 (mCripto-1) expression that occurs during mammary gland development and a stage-specific function of mCripto-1 signaling during mammary gland development. Our data suggest that Cripto-1, like the progesterone receptor, is not required for the initial ductal growth but is essential for subsequent side branching and alveologenesis during the initial stages of pregnancy. Dissection of the mechanism by which this occurs indicates that mCripto-1 activates receptor activator NF-κB/receptor activator NF-κB ligand, and NF-κB signaling pathways.


Subject(s)
Epidermal Growth Factor/metabolism , Membrane Glycoproteins/metabolism , NF-kappa B p50 Subunit/metabolism , Neoplasm Proteins/metabolism , RANK Ligand/metabolism , Receptor Activator of Nuclear Factor-kappa B/metabolism , Receptors, Progesterone/metabolism , Signal Transduction , Animals , Cell Proliferation , Endoplasmic Reticulum Chaperone BiP , Epidermal Growth Factor/genetics , Epithelial Cells , Epithelial-Mesenchymal Transition , Female , Humans , Mammary Glands, Animal/cytology , Membrane Glycoproteins/genetics , Mice , Mice, Inbred BALB C , Mice, Knockout , Models, Biological , NF-kappa B p50 Subunit/genetics , Neoplasm Proteins/genetics , Organ Specificity , Pregnancy , RANK Ligand/genetics , Receptor Activator of Nuclear Factor-kappa B/genetics , Receptors, Progesterone/genetics
2.
Connect Tissue Res ; 56(5): 364-80, 2015.
Article in English | MEDLINE | ID: mdl-26327334

ABSTRACT

Cripto-1 (CR-1) is a multifunctional embryonic protein that is re-expressed during inflammation, wound repair, and malignant transformation. CR-1 can function either as a tethered co-receptor or shed as a free ligand underpinning its flexible role in cell physiology. CR-1 has been shown to mediate cell growth, migration, invasion, and induce epithelial to mesenchymal transition (EMT). The main signaling pathways mediating CR-1 effects include Nodal-dependent (Smad2/3) and Nodal-independent (Src/p44/42/Akt) signaling transduction pathways. In addition, there are several naturally occurring binding partner proteins (BPPs) for CR-1 that can either agonize or antagonize its bioactivity. We will review the collective role of CR-1 as an extracellular protein, discuss caveats to consider in developing a quantitation assay, define possible mechanistic avenues applicable for drug discovery, and report on our experimental approaches to overcome these problematic issues.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , GPI-Linked Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Signal Transduction/physiology , Autoantibodies/immunology , Epidermal Growth Factor/physiology , Epithelial-Mesenchymal Transition/immunology , Extracellular Space/metabolism , Humans , Signal Transduction/immunology , Transforming Growth Factor beta/metabolism
3.
Minerva Med ; 115(1): 68-82, 2024 Feb.
Article in English | MEDLINE | ID: mdl-37947781

ABSTRACT

Sleep-disordered breathing has a relatively high prevalence, which varies from 3-7% in males and from 2-5% in females in the adult population. Studies published in the literature have shown that sleep apnea is closely related to an increased risk of developing various pathologies, among which arterial hypertension stands out. The prevalence of hypertension in patients suffering from obstructive sleep apnea (OSA) ranges from 35-80% and appears to be related to OSA severity. Approximately 40-50% of patients affected by hypertension are also affected by OSA and this association seems to be stronger in young and middle-aged adults (<50 years of age). The primary objective of this narrative review is to provide an update on what are the main contributing comorbidities to the development of a hypertensive state in patients suffering from OSA, an independent risk factor for diurnal hypertension, implicated as a risk factor for the first stroke, recurrent stroke, and post-stroke mortality. There are a lot of factors that contribute to developing a hypertensive state in OSA patients, some more decisive, others less. More evidence from longitudinal studies is needed on the impact of OSA on cardiovascular risk in females, on the causal link between OSA and arterial hypertension or metabolic diseases, like diabetes and glucose intolerance, and the effect of different kinds of OSA treatment.


Subject(s)
Diabetes Mellitus , Hypertension , Sleep Apnea, Obstructive , Adult , Female , Humans , Male , Middle Aged , Comorbidity , Diabetes Mellitus/epidemiology , Hypertension/complications , Hypertension/epidemiology , Risk Factors , Sleep Apnea, Obstructive/complications , Sleep Apnea, Obstructive/epidemiology , Young Adult
4.
Clin Transl Allergy ; 14(1): e12325, 2024 Jan.
Article in English | MEDLINE | ID: mdl-38282193

ABSTRACT

INTRODUCTION: Information and communication technologies (ICTs) improve patient-centered care and are routinely used in Allergic Rhinitis (AR), but patients' preferences and attitudes are unexplored. This study examines AR-related information preferences and ICT use by AR patients. METHODS: A survey-based cross-sectional study was carried out in Ecuador from July to September 2019 in seven centers of reference for allergic disease. Participants were 18 years or older, diagnosed with AR and had access to ICT and the Internet. Descriptive and binomial logistic regressions were performed. A value of less than 0.05 was considered statistically significant. RESULTS: 217 patients were included. 47% (n = 102) used ICTs to learn about AR, of which 38.2% (n = 83) found it useful. Most of participants (75%, n = 164) did not think that ICTs reduce their need to see a doctor. Individuals with poorer quality of life were more likely to utilize ICTs to contact their doctor (OR 1.27, 95% CI 1.04-1.55), and more likely to be interested in AR-related content (OR 1.23, 95% CI 1.00-1.52). Patients with long-term AR or other allergies were less likely to use ICTs (OR 0.92 and OR 0.40 respectively). Higher education and lower quality of life may increase AR apps adoption (OR 4.82, 95% CI 1.11-21.00). Academic preparation five-fold increased ICT use for health provider communication (OR 5.29, 95% CI 1.18-23.72). Mild-persistent AR enhanced the probabilities of using ICTs to share experiences and communicate with other patients (OR 12.59, 95% CI 1.32-120.35). CONCLUSIONS: Our study emphasizes the importance of tailoring digital resources to patient needs by considering factors such as quality of life, education, and specific subgroups within the AR patient population. Additionally, the findings suggest that while ICTs can play a valuable role in patient education and support, they should complement, rather than replace, traditional medical care for many AR patients.

5.
Minerva Med ; 115(1): 4-13, 2024 Feb.
Article in English | MEDLINE | ID: mdl-38261298

ABSTRACT

BACKGROUND: Chest physiotherapy plays a crucial role in the treatment of COPD, although the optimal techniques for airway clearance have not been definitively established. Among the different techniques, high-frequency chest wall oscillation (HFCWO) has gained attention for its potential to create a widespread lung percussion, facilitating the removal of secretions and potentially clearing the peripheral bronchial tree. This study aims to assess the effectiveness of a novel "focused pulse" HFCWO in patients with moderate to severe COPD. METHODS: Sixty patients were randomized to three groups: a group treated with the PEP technique, a group with "focused pulse "HFCWO" and a group with pharmacological therapy alone (control group). The primary outcomes were changes in respiratory function parameters, changes in dyspnea and quality of life scores as well as daily life activity and health status assessment. The secondary outcomes were the number of exacerbations and the number of practitioner or emergency department (ED) visits after 1, 3, and 6 months. RESULTS: Sixty patients concluded the study with 20 patients allocated to each group. The two devices improved respiratory function tests, quality of life and health scores and dyspnea compared to the control group. Maximal expiratory pressure and diffusing lung carbon oxide were significantly improved in the focused pulse HFCWO group compared to the PEP group. Only pulse-focused HFCWO showed a statistically significant lower number of exacerbations and visits to ED or practitioner compared to the control group. CONCLUSIONS: The focused pulse HFCWO technique improves daily life activities and lung function in patients with stable COPD. The device demonstrated significantly greater effectiveness in lowering COPD exacerbations as well as visits to ED or practitioner.


Subject(s)
Chest Wall Oscillation , Pulmonary Disease, Chronic Obstructive , Humans , Chest Wall Oscillation/methods , Quality of Life , Lung , Pulmonary Disease, Chronic Obstructive/therapy , Dyspnea/etiology , Dyspnea/therapy
6.
J Cell Physiol ; 228(6): 1174-88, 2013 Jun.
Article in English | MEDLINE | ID: mdl-23129342

ABSTRACT

Human Cripto-1 (CR-1) plays an important role in regulating embryonic development while also regulating various stages of tumor progression. However, mechanisms that regulate CR-1 expression during embryogenesis and tumorigenesis are still not well defined. In the present study, we investigated the effects of two nuclear receptors, liver receptor homolog (LRH)-1 and germ cell nuclear factor receptor (GCNF) and epigenetic modifications on CR-1 gene expression in NTERA-2 human embryonal carcinoma cells and in breast cancer cells. CR-1 expression in NTERA-2 cells was positively regulated by LRH-1 through direct binding to a DR0 element within the CR-1 promoter, while GCNF strongly suppressed CR-1 expression in these cells. In addition, the CR-1 promoter was unmethylated in NTERA-2 cells, while T47D, ZR75-1, and MCF7 breast cancer cells showed high levels of CR-1 promoter methylation and low CR-1 mRNA and protein expression. Treatment of breast cancer cells with a demethylating agent and histone deacetylase inhibitors reduced methylation of the CR-1 promoter and reactivated CR-1 mRNA and protein expression in these cells, promoting migration and invasion of breast cancer cells. Analysis of a breast cancer tissue array revealed that CR-1 was highly expressed in the majority of human breast tumors, suggesting that CR-1 expression in breast cancer cell lines might not be representative of in vivo expression. Collectively, these findings offer some insight into the transcriptional regulation of CR-1 gene expression and its critical role in the pathogenesis of human cancer.


Subject(s)
Breast Neoplasms/metabolism , Carcinoma, Ductal, Breast/metabolism , Carcinoma, Embryonal/metabolism , DNA Methylation , GPI-Linked Proteins/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Neoplasm Proteins/metabolism , Nuclear Receptor Subfamily 6, Group A, Member 1/metabolism , Promoter Regions, Genetic , Receptors, Cytoplasmic and Nuclear/metabolism , Azacitidine/analogs & derivatives , Azacitidine/pharmacology , Binding Sites , Breast Neoplasms/genetics , Breast Neoplasms/pathology , Carcinoma, Ductal, Breast/genetics , Carcinoma, Ductal, Breast/pathology , Carcinoma, Embryonal/genetics , Carcinoma, Embryonal/pathology , Cell Movement , DNA Methylation/drug effects , DNA Modification Methylases/antagonists & inhibitors , DNA Modification Methylases/metabolism , Decitabine , Dose-Response Relationship, Drug , Embryonal Carcinoma Stem Cells/metabolism , Embryonal Carcinoma Stem Cells/pathology , Female , GPI-Linked Proteins/genetics , Gene Expression Regulation, Developmental , Gene Expression Regulation, Neoplastic , Genes, Reporter , Histone Deacetylase Inhibitors/pharmacology , Humans , Hydroxamic Acids/pharmacology , Intercellular Signaling Peptides and Proteins/genetics , Luciferases/biosynthesis , Luciferases/genetics , MCF-7 Cells , Neoplasm Invasiveness , Neoplasm Proteins/genetics , Nuclear Receptor Subfamily 6, Group A, Member 1/genetics , RNA Interference , RNA, Messenger/metabolism , Receptors, Cytoplasmic and Nuclear/genetics , Time Factors , Tissue Array Analysis , Transcription, Genetic , Transfection , Tretinoin/pharmacology , Valproic Acid/pharmacology
7.
J Hematol Oncol ; 15(1): 116, 2022 08 26.
Article in English | MEDLINE | ID: mdl-36028857

ABSTRACT

BACKGROUND: Patients with chronic lymphocytic leukemia (CLL) may be more susceptible to COVID-19 related poor outcomes, including thrombosis and death, due to the advanced age, the presence of comorbidities, and the disease and treatment-related immune deficiency. The aim of this study was to assess the risk of thrombosis and bleeding in patients with CLL affected by severe COVID-19. METHODS: This is a retrospective multicenter study conducted by ERIC, the European Research Initiative on CLL, including patients from 79 centers across 22 countries. Data collection was conducted between April and May 2021. The COVID-19 diagnosis was confirmed by the real-time polymerase chain reaction (RT-PCR) assay for SARS-CoV-2 on nasal or pharyngeal swabs. Severe cases of COVID-19 were defined by hospitalization and the need of oxygen or admission into ICU. Development and type of thrombotic events, presence and severity of bleeding complications were reported during treatment for COVID-19. Bleeding events were classified using ISTH definition. STROBE recommendations were used in order to enhance reporting. RESULTS: A total of 793 patients from 79 centers were included in the study with 593 being hospitalized (74.8%). Among these, 511 were defined as having severe COVID: 162 were admitted to the ICU while 349 received oxygen supplementation outside the ICU. Most patients (90.5%) were receiving thromboprophylaxis. During COVID-19 treatment, 11.1% developed a thromboembolic event, while 5.0% experienced bleeding. Thrombosis developed in 21.6% of patients who were not receiving thromboprophylaxis, in contrast to 10.6% of patients who were on thromboprophylaxis. Bleeding episodes were more frequent in patients receiving intermediate/therapeutic versus prophylactic doses of low-molecular-weight heparin (LWMH) (8.1% vs. 3.8%, respectively) and in elderly. In multivariate analysis, peak D-dimer level and C-reactive protein to albumin ratio were poor prognostic factors for thrombosis occurrence (OR = 1.022, 95%CI 1.007‒1.038 and OR = 1.025, 95%CI 1.001‒1.051, respectively), while thromboprophylaxis use was protective (OR = 0.199, 95%CI 0.061‒0.645). Age and LMWH intermediate/therapeutic dose administration were prognostic factors in multivariate model for bleeding (OR = 1.062, 95%CI 1.017-1.109 and OR = 2.438, 95%CI 1.023-5.813, respectively). CONCLUSIONS: Patients with CLL affected by severe COVID-19 are at a high risk of thrombosis if thromboprophylaxis is not used, but also at increased risk of bleeding under the LMWH intermediate/therapeutic dose administration.


Subject(s)
COVID-19 Drug Treatment , Leukemia, Lymphocytic, Chronic, B-Cell , Thrombosis , Venous Thromboembolism , Aged , Anticoagulants , COVID-19 Testing , Hemorrhage , Heparin, Low-Molecular-Weight , Humans , SARS-CoV-2
8.
Am J Pathol ; 177(2): 532-40, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20616345

ABSTRACT

Cripto-1 is critical for early embryonic development and, together with its ligand Nodal, has been found to be associated with the undifferentiated status of mouse and human embryonic stem cells. Like other embryonic genes, Cripto-1 performs important roles in the formation and progression of several types of human tumors, stimulating cell proliferation, migration, epithelial to mesenchymal transition, and tumor angiogenesis. Several studies have demonstrated that cell fate regulation during embryonic development and cell transformation during oncogenesis share common signaling pathways, suggesting that uncontrolled activation of embryonic signaling pathways might drive cell transformation and tumor progression in adult tissues. Here we review our current understanding of how Cripto-1 controls stem cell biology and how it integrates with other major embryonic signaling pathways. Because many cancers are thought to derive from a subpopulation of cancer stem-like cells, which may re-express embryonic genes, Cripto-1 signaling may drive tumor growth through the generation or expansion of tumor initiating cells bearing stem-like characteristics. Therefore, the Cripto-1/Nodal signaling may represent an attractive target for treatment in cancer, leading to the elimination of undifferentiated stem-like tumor initiating cells.


Subject(s)
Disease Progression , Epidermal Growth Factor/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Neoplasms/pathology , Stem Cells/physiology , Animals , Embryonic Development , Epidermal Growth Factor/genetics , Epithelial-Mesenchymal Transition , GPI-Linked Proteins , Humans , Hypoxia , Intercellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Neoplasm Proteins/genetics , Neoplastic Stem Cells/metabolism , Neoplastic Stem Cells/pathology , Nodal Protein/genetics , Nodal Protein/metabolism , Receptors, Notch/genetics , Receptors, Notch/metabolism , Signal Transduction/physiology , Wnt Proteins/genetics , Wnt Proteins/metabolism
9.
Stem Cells ; 28(8): 1303-14, 2010 Aug.
Article in English | MEDLINE | ID: mdl-20549704

ABSTRACT

Deregulation of stem cells is associated with the generation and progression of malignant tumors. In addition, genes that are associated with early embryogenesis are frequently expressed in cancer. Cripto-1 (CR-1), a glycosylphosphatidylinositol-linked glycoprotein, is expressed during early embryogenesis and in various human carcinomas. We demonstrated that human embryonal carcinoma (EC) cells are heterogeneous for CR-1 expression and consist of two distinct subpopulations: a CR-1(High) and a CR-1(Low) population. By segregating CR-1(High) and CR-1(Low) populations of NTERA2/D1 EC cells by fluorescence-activated cell sorting, we demonstrated that CR-1(High) cells were more tumorigenic than CR-1(Low) cells by an in vitro tumor sphere assay and by in vivo xenograft formation. The CR-1(High) population was enriched in mRNA expression for the pluripotent embryonic stem (ES) cell genes Oct4, Sox2, and Nanog. CR-1 expression in NTERA2/D1 cells was regulated by a Smad2/3-dependent autocrine loop, by the ES cell-related transcription factors Oct4/Nanog, and partially by the DNA methylation status of the promoter region. These results demonstrate that CR-1 expression is enriched in an undifferentiated, tumorigenic subpopulation and is regulated by key regulators of pluripotent stem cells.


Subject(s)
Embryonal Carcinoma Stem Cells/cytology , Embryonal Carcinoma Stem Cells/metabolism , Epidermal Growth Factor/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Animals , Blotting, Western , Cell Line , Chromatin Immunoprecipitation , DNA Methylation , Epidermal Growth Factor/genetics , Flow Cytometry , Fluorescent Antibody Technique , GPI-Linked Proteins , Homeodomain Proteins/genetics , Homeodomain Proteins/metabolism , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins , Membrane Glycoproteins/genetics , Mice , Mice, Nude , Nanog Homeobox Protein , Neoplasm Proteins/genetics , Neoplasm Transplantation , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pluripotent Stem Cells/cytology , Pluripotent Stem Cells/metabolism , Reverse Transcriptase Polymerase Chain Reaction
10.
Am J Pathol ; 175(5): 2146-58, 2009 Nov.
Article in English | MEDLINE | ID: mdl-19834060

ABSTRACT

Cripto-1 is a membrane-bound protein that is highly expressed in embryonic stem cells and in human tumors. In the present study, we investigated the effect of low levels of oxygen, which occurs naturally in rapidly growing tissues, on Cripto-1 expression in mouse embryonic stem (mES) cells and in human embryonal carcinoma cells. During hypoxia, Cripto-1 expression levels were significantly elevated in mES cells and in Ntera-2 or NCCIT human embryonal carcinoma cells, as compared with cells growing with normal oxygen levels. The transcription factor hypoxia-inducible factor-1alpha directly regulated Cripto-1 expression by binding to hypoxia-responsive elements within the promoter of mouse and human Cripto-1 genes in mES and NCCIT cells, respectively. Furthermore, hypoxia modulated differentiation of mES cells by enhancing formation of beating cardiomyocytes as compared with mES cells that were differentiated under normoxia. However, hypoxia failed to induce differentiation of mES cells into cardiomyocytes in the absence of Cripto-1 expression, demonstrating that Cripto-1 is required for hypoxia to fully differentiate mES cells into cardiomyocytes. Finally, cardiac tissue samples derived from patients who had suffered ischemic heart disease showed a dramatic increase in Cripto-1 expression as compared with nonischemic heart tissue samples, suggesting that hypoxia may also regulate Cripto-1 in vivo.


Subject(s)
Cell Differentiation/physiology , Embryonic Stem Cells/physiology , Epidermal Growth Factor/metabolism , Heart , Hypoxia/metabolism , Membrane Glycoproteins/metabolism , Myocytes, Cardiac/physiology , Neoplasm Proteins/metabolism , Animals , Biomarkers/metabolism , Cell Line , Embryonic Stem Cells/cytology , Epidermal Growth Factor/genetics , Heart/anatomy & histology , Heart/embryology , Humans , Hypoxia-Inducible Factor 1, alpha Subunit/genetics , Hypoxia-Inducible Factor 1, alpha Subunit/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Knockout , Myocardium/cytology , Myocardium/metabolism , Myocytes, Cardiac/cytology , Neoplasm Proteins/genetics , Promoter Regions, Genetic , RNA, Small Interfering/genetics , RNA, Small Interfering/metabolism , Response Elements , Signal Transduction/physiology , Swine
11.
Biochim Biophys Acta ; 1778(12): 2671-81, 2008 Dec.
Article in English | MEDLINE | ID: mdl-18930707

ABSTRACT

Epidermal Growth Factor-Cripto-1/FRL-1/Cryptic (EGF-CFC) proteins, including human Cripto-1 (hCFC2/hCR-1) and human Cryptic (hCFC1), are membrane-associated Nodal co-receptors, which have critical roles in vertebrate development. Most of the EGF-CFC proteins have been experimentally proven or predicted to be glycosylphosphatidylinositol (GPI)-anchored proteins. However, unlike other EGF-CFC proteins, hCFC1 does not exhibit a typical GPI-signal sequence, containing a 32-amino acid hydrophilic extension in its COOH-terminal end. Here we experimentally demonstrate that the COOH-terminal sequence of hCFC1 functions as a GPI-anchoring signal. Moreover, addition of a hydrophilic epitope tag of 55-amino acids (V5-His) after the GPI signal of hCR-1 interfered with generation of a GPI-anchored form of hCR-1. In contrast, addition of the same epitope tag to the end of GPI signal of hCFC1 did not affect the GPI-attachment of hCFC1. The COOH-terminal signal of hCFC1 could produce two different forms of the protein; a GPI-anchored form and an unprocessed form which was more prone to be secreted into the conditioned medium. The hydrophilic extension of hCFC1 negatively regulates the activity of hCFC1 as a Nodal co-receptor. These results demonstrate the presence of endogenous GPI-signal sequence with a hydrophilic extension, which can generate both GPI-anchored and soluble forms of the protein.


Subject(s)
Epidermal Growth Factor/metabolism , Glycosylphosphatidylinositols/metabolism , Intercellular Signaling Peptides and Proteins/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Nodal Protein/metabolism , Protein Sorting Signals/physiology , Amino Acid Sequence , Cell Line , Epidermal Growth Factor/genetics , GPI-Linked Proteins , Genes, Reporter , Glycosylphosphatidylinositols/genetics , Humans , Immunohistochemistry , Intercellular Signaling Peptides and Proteins/genetics , Kidney/cytology , Luciferases/metabolism , Membrane Glycoproteins/genetics , Molecular Sequence Data , Neoplasm Proteins/genetics , Nodal Protein/genetics , Protein Sorting Signals/genetics , Transfection
12.
Cell Signal ; 20(9): 1632-41, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18595660

ABSTRACT

Both canonical Wnt/beta-catenin and TGFbeta/Smad signaling pathways coordinately regulate pattern formation during embryogenesis as well as tumor progression. Evidence of cross-talk between these two pathways has been reported. Here we demonstrated that the Activin-like kinase 4 (Alk4)/Smad2 pathway facilitates the transcriptional activity of the oncogenic Wnt/beta-catenin/Tcf4 pathway through a novel Smad4-independent mechanism. Upon activation, Smad2 physically interacted with Tcf4, beta-catenin and the co-activator p300 to enhance transcriptional activity of beta-catenin/Tcf4 through the histone acetyltransferase activity of p300. Transactivation by Smad2 was independent of a Smad-binding element (SBE) and Smad4. Indeed, the enhancement of beta-catenin/Tcf4 transcriptional activity by activated Smad2 was negatively regulated by the presence of Smad4. Moreover, a tumor-derived missense mutant of Smad2, lacking the ability to bind to Smad4 was still able to enhance the Tcf4 transcriptional reporter in the presence of beta-catenin and Tcf4. Our findings suggest that Smad2 may function as an activator of canonical Wnt/beta-catenin/Tcf4 signaling through a SBE/Smad4-independent pathway.


Subject(s)
Histone Acetyltransferases/metabolism , Signal Transduction , Smad2 Protein/metabolism , Wnt Proteins/metabolism , beta Catenin/metabolism , p300-CBP Transcription Factors/metabolism , Activin Receptors, Type I/metabolism , Cell Line , Humans , Models, Genetic , Protein Binding , Smad2 Protein/genetics , Smad4 Protein/metabolism , TCF Transcription Factors/metabolism , Transcription Factor 7-Like 2 Protein , Transcription, Genetic , Transcriptional Activation
13.
J Cell Physiol ; 215(1): 192-203, 2008 Apr.
Article in English | MEDLINE | ID: mdl-17941089

ABSTRACT

Human Cripto-1 (CR-1) is a cell membrane protein that is overexpressed in several different types of human carcinomas. In the present study we investigated the mechanisms that regulate the expression of CR-1 gene in cancer cells. We cloned a 2,481 bp 5'-flanking region of the human CR-1 gene into a luciferase reporter vector and transfected NTERA-2 human embryonal carcinoma cells and LS174-T colon cancer cells to test for promoter activity. Activity of CR-1 promoter in both cell lines was modulated by two TGF-beta family members, TGF-beta1 and BMP-4. In particular, TGF-beta1 significantly up-regulated CR-1 promoter activity, whereas a dramatic reduction in CR-1 promoter activity was observed with BMP-4 in NTERA-2 and LS174-T cells. Changes in the CR-1 promoter activity following TGF-beta1 and BMP-4 treatments correlated with changes in CR-1 mRNA and protein expression in NTERA-2 and LS174-T cells. We also identified three Smad binding elements (SBEs) within the CR-1 promoter and point mutation of SBE1 (-2,197/-2,189) significantly reduced response of the CR-1 promoter to both TGF-beta1 and BMP-4 in NTERA-2 and LS174-T cells. Chromatin immunoprecipitation assay also demonstrated binding of Smad-4 to a CR-1 promoter DNA sequence containing SBE1 in LS174-T cells. Finally, BMP-4 inhibited migration of LS174-T cells and F9 mouse embryonal carcinoma cells by downregulation of CR-1 protein. In conclusion, these results suggest a differential modulation of CR-1 gene expression in embryonal and colon cancer cells by two different members of the TGF-beta family.


Subject(s)
Bone Morphogenetic Proteins/pharmacology , Colonic Neoplasms/genetics , Embryonal Carcinoma Stem Cells/metabolism , Epidermal Growth Factor/genetics , Gene Expression Regulation, Neoplastic/drug effects , Membrane Glycoproteins/genetics , Neoplasm Proteins/genetics , Transforming Growth Factor beta1/pharmacology , 5' Flanking Region , Animals , Base Sequence , Bone Morphogenetic Protein 4 , Cell Movement/drug effects , Cell Proliferation/drug effects , Colonic Neoplasms/pathology , Embryonal Carcinoma Stem Cells/pathology , Epidermal Growth Factor/deficiency , Epidermal Growth Factor/metabolism , GPI-Linked Proteins , Humans , Intercellular Signaling Peptides and Proteins , Membrane Glycoproteins/deficiency , Membrane Glycoproteins/metabolism , Mice , Molecular Sequence Data , Mutation/genetics , Neoplasm Proteins/deficiency , Neoplasm Proteins/metabolism , Promoter Regions, Genetic , Protein Binding/drug effects , RNA, Messenger/genetics , RNA, Messenger/metabolism , Smad Proteins/metabolism
14.
J Cell Physiol ; 216(3): 824-34, 2008 Sep.
Article in English | MEDLINE | ID: mdl-18425773

ABSTRACT

Netrin-1 has been shown to regulate the function of the EGF-like protein Cripto-1 (Cr-1) and affect mammary gland development. Since Cr-1 is a target gene of Nanog and Oct4, we investigated the relationship between Netrin-1 and Cr-1, Nanog and Oct4 during different stages of development in the mouse mammary gland. Results from histological analysis show that exogenous Netrin-1 was able to induce formation of alveolar-like structures within the mammary gland terminal end buds of virgin transgenic Cripto-1 mice and enhance mammary gland alveologenesis in early pregnant FVB/N mice. Results from immunostaining and Western blot analysis show that Netrin-1, Nanog and Oct4 are expressed in the mouse embryonic mammary anlage epithelium while Cripto-1 is predominantly expressed outside this structure in the surrounding mesenchyme. We find that in lactating mammary glands of postnatal FVB/N mice, Netrin-1 expression is highest while Cripto-1 and Nanog levels are lowest indicating that Netrin-1 may perform a role in the mammary gland during lactation. HC-11 mouse mammary epithelial cells stimulated with lactogenic hormones and exogenous soluble Netrin-1 showed increased beta-casein expression as compared to control thus supporting the potential role for Netrin-1 during functional differentiation of mouse mammary epithelial cells. Finally, mouse ES cells treated with exogenous soluble Netrin-1 showed reduced levels of Nanog and Cripto-1 and higher levels of beta-III tubulin during differentiation. These results suggest that Netrin-1 may facilitate functional differentiation of mammary epithelial cells and possibly affect the expression of Nanog and/or Cripto-1 in multipotent cells that may reside in the mammary gland.


Subject(s)
Epidermal Growth Factor/metabolism , Homeodomain Proteins/metabolism , Mammary Glands, Animal/growth & development , Membrane Glycoproteins/metabolism , Morphogenesis/physiology , Neoplasm Proteins/metabolism , Nerve Growth Factors/metabolism , Tumor Suppressor Proteins/metabolism , Animals , Caseins/metabolism , Cell Differentiation , Cells, Cultured , Dexamethasone/metabolism , Embryonic Stem Cells/cytology , Epidermal Growth Factor/genetics , Female , Gene Expression Regulation, Developmental , Glucocorticoids/metabolism , Homeodomain Proteins/genetics , Insulin/metabolism , Lactation , Male , Mammary Glands, Animal/anatomy & histology , Mammary Glands, Animal/metabolism , Membrane Glycoproteins/genetics , Mice , Mice, Transgenic , Nanog Homeobox Protein , Neoplasm Proteins/genetics , Nerve Growth Factors/genetics , Netrin-1 , Octamer Transcription Factor-3/genetics , Octamer Transcription Factor-3/metabolism , Pregnancy , Prolactin/metabolism , Tumor Suppressor Proteins/genetics
15.
FEBS Lett ; 582(29): 3997-4002, 2008 Dec 10.
Article in English | MEDLINE | ID: mdl-19013461

ABSTRACT

Cripto-1, a co-receptor for Nodal, can activate Nodal-dependent and Nodal-independent signaling pathways. In this study we have investigated whether Cripto-1 mutants, that fail to activate a Nodal-dependent signaling pathway, are capable to activate a Nodal-independent signaling pathway in mammary epithelial cells. Cripto-1 mutants expressed in EpH4 mouse mammary epithelial cells are fully functional in regard to activation of a Nodal-independent signaling pathway, leading to phosphorylation of mitogen-activated protein kinase (MAPK) and Akt and to enhanced proliferation and motility of these cells, suggesting that Cripto-1 mutants with impaired Nodal signaling are still active in a Nodal-independent signaling pathway.


Subject(s)
Epidermal Growth Factor/metabolism , Membrane Glycoproteins/metabolism , Neoplasm Proteins/metabolism , Nodal Protein/metabolism , Animals , COS Cells , Cell Line , Cell Movement , Cell Proliferation , Chlorocebus aethiops , Epidermal Growth Factor/genetics , Epithelial Cells/metabolism , Glypicans/metabolism , Mammary Glands, Animal , Membrane Glycoproteins/genetics , Mice , Neoplasm Proteins/genetics , Signal Transduction
16.
Clin Cancer Res ; 12(8): 2526-37, 2006 Apr 15.
Article in English | MEDLINE | ID: mdl-16638862

ABSTRACT

PURPOSE: Two clinical trials were conducted to evaluate the clinical efficacy and immunologic impact of vaccination against the tyrosinase protein plus systemic interleukin 2 (IL-2) administration in patients with advanced metastatic melanoma. EXPERIMENTAL DESIGN: Full-length tyrosinase was employed as an immunogen to induce diverse immunologic responses against a commonly expressed melanoma antigen. Heterologous prime/boost vaccination with recombinant vaccinia and fowlpox vectors encoding tyrosinase was first explored in a randomized three-arm phase II trial, in which vaccines were administered alone or concurrently with low-dose or high-dose IL-2. In a subsequent single cohort phase II trial, all patients received the same vaccines and high-dose IL-2 sequentially rather than concurrently. RESULTS: Among a total of 64 patients treated on these trials, 8 objective partial responses (12.5%) were observed, all in patients receiving high-dose IL-2. Additional patients showed evidence of lesional regression (mixed tumor response) or overall regression that did not achieve partial response status (minor response). In vitro evidence of enhanced immunity against tyrosinase following protocol treatments was documented in 3 of 49 (6%) patients tested serologically, 3 of 23 (13%) patients tested for T-cell recognition of individual tyrosinase peptides, and 4 of 16 (25%) patients tested for T-cell recognition of full-length tyrosinase protein with real-time reverse transcription-PCR techniques. CONCLUSIONS: Whereas prime/boost immunization with recombinant vaccinia and fowlpox viruses enhanced antityrosinase immunity in some patients with metastatic melanoma, it was ineffective alone in mediating clinical benefit, and in combination with IL-2 did not mediate clinical benefit significantly different from that expected from treatment with IL-2 alone.


Subject(s)
Cancer Vaccines/immunology , DNA, Recombinant/immunology , Immunization, Secondary/methods , Interleukin-2/therapeutic use , Melanoma/therapy , Vaccination/methods , Combined Modality Therapy , DNA, Recombinant/genetics , Genetic Vectors/genetics , Humans , Immunization Schedule , Immunoglobulin G/blood , Interferon-gamma/genetics , Interferon-gamma/metabolism , Interleukin-2/administration & dosage , Melanoma/immunology , Melanoma/pathology , Monophenol Monooxygenase/genetics , Monophenol Monooxygenase/immunology , Monophenol Monooxygenase/metabolism , Neoplasm Metastasis , Poxviridae/genetics , RNA, Messenger/genetics , RNA, Messenger/metabolism , Reverse Transcriptase Polymerase Chain Reaction , T-Lymphocytes/drug effects , T-Lymphocytes/immunology , T-Lymphocytes/metabolism , Treatment Outcome
17.
Cancer Res ; 64(5): 1595-9, 2004 Mar 01.
Article in English | MEDLINE | ID: mdl-14996715

ABSTRACT

The potential of antigen-directed cancer immunotherapy has not been fully realized, perhaps because many commonly targeted tumor associated proteins are not essential to maintaining the malignant cell phenotype. A constitutively activating mutation in the signaling molecule BRAF is expressed frequently in melanomas and may play an important role in the biology of this disease. A 29-mer B-Raf peptide incorporating the V599E mutation was used for in vitro stimulation of lymphocytes derived from melanoma patients, generating MHC class II-restricted CD4(+) T cells specific for this peptide as well as for melanoma cells expressing B-Raf V599E. Mutated B-Raf exemplifies targets that may be ideal for immunotherapy.


Subject(s)
CD4-Positive T-Lymphocytes/immunology , Melanoma/genetics , Mutation , Proto-Oncogene Proteins c-raf/genetics , Alleles , Amino Acid Sequence , HLA-DP Antigens/genetics , HLA-DP beta-Chains , HLA-DQ Antigens/genetics , HLA-DQ beta-Chains , HLA-DR Antigens/genetics , HLA-DRB1 Chains , Humans , Melanoma/immunology , Molecular Sequence Data , Proto-Oncogene Proteins B-raf , Proto-Oncogene Proteins c-raf/chemistry , Proto-Oncogene Proteins c-raf/immunology , Sequence Analysis, DNA
18.
Cancer Res ; 62(19): 5505-9, 2002 Oct 01.
Article in English | MEDLINE | ID: mdl-12359760

ABSTRACT

CD4+ T-cell responses against human tumor antigens are a potentially critical component of the antitumor immune response. Molecular methods have been devised for rapidly identifying MHC class II-restricted tumor antigens and elucidating the recognized epitopes. We describe here the identification of neo-poly(A) polymerase (neo-PAP), a novel RNA processing enzyme overexpressed in a variety of human cancers, by screening a melanoma-derived invariant chain fusion cDNA library with tumor-reactive CD4+ T lymphocytes. A cryptic nonmutated HLA-DRbeta1*0701-restricted neo-PAP epitope was processed through the endogenous MHC class II pathway. A unique point mutation effected a nonconservative substitution of a leucine for a proline residue at a structurally important site in neo-PAP that was remote from the recognized peptide, revealing a normally silent epitope for immune recognition. Genetic aberrations such as the described point mutation can have unexpected immunological consequences, in this case leading to immune recognition of a distant normal self epitope.


Subject(s)
Epitopes, T-Lymphocyte/immunology , HLA-DR7 Antigen/immunology , Melanoma/immunology , Polynucleotide Adenylyltransferase/immunology , RNA, Neoplasm/metabolism , Adult , Alleles , Amino Acid Sequence , Antigens, Neoplasm/immunology , CD4-Positive T-Lymphocytes/immunology , Epitopes, T-Lymphocyte/genetics , Gene Library , Humans , Male , Melanoma/enzymology , Molecular Sequence Data , Point Mutation , Polynucleotide Adenylyltransferase/genetics , Polynucleotide Adenylyltransferase/metabolism , Subcellular Fractions/enzymology
19.
Hum Gene Ther ; 15(4): 393-404, 2004 Apr.
Article in English | MEDLINE | ID: mdl-15053864

ABSTRACT

Directing the human immune system to recognize and eliminate tumor cells is the ultimate goal of cancer immunotherapy. Vaccinating patients with autologous antigen presenting cells (APC) expressing tumor-associated antigens (TAA) represents a promising approach for activating tumor-reactive T cells in vivo. In addition, APC expressing TAA provide a means of generating tumor-specific T cells in vitro, for therapeutic and diagnostic applications. Lentiviral vectors are attractive vehicles for introducing TAA-encoding genes into APC. In this study, lentiviral vectors expressing the reporter gene GFP or the melanoma-associated antigen tyrosinase were used to transduce three different kinds of human APC: monocyte-derived dendritic cells (DC), CD40L-activated B lymphocytes, and Epstein Barr virus (EBV)-transformed B lymphocytes. Using optimized transduction conditions for each cell type, tyrosinase was expressed at levels sufficient to stimulate antigen-specific major histocompatibility complex (MHC) class I-restricted T cells from melanoma patients. While transduced EBV-B cells demonstrated the highest level of transgene expression, optimal T-cell recognition was achieved with transduced DC. Substituting the CAG promoter for PGK in lentiviral constructs enhanced transgene expression in DC and EBV-B cells, amplifying T cell recognition. Lentiviruses inducing sustained transgene expression with relatively low cellular toxicity and background viral gene expression may be ideal vectors for immunotherapeutic applications.


Subject(s)
Antigen-Presenting Cells/immunology , Antigens, Neoplasm/immunology , Dendritic Cells/physiology , Epitopes , Genetic Vectors , Lentivirus/genetics , Antigens, Neoplasm/metabolism , B-Lymphocytes/immunology , CD40 Ligand/metabolism , Cell Transformation, Viral , Dendritic Cells/immunology , Dendritic Cells/virology , Gene Transfer Techniques , Genes, MHC Class I , Green Fluorescent Proteins , HeLa Cells , Herpesvirus 4, Human/genetics , Humans , Luminescent Proteins/metabolism , Melanoma/genetics , Melanoma/pathology , Melanoma-Specific Antigens , Monocytes/cytology , Monocytes/virology , Monophenol Monooxygenase/metabolism , Neoplasm Proteins/metabolism , Promoter Regions, Genetic/genetics , T-Lymphocytes/immunology , Transduction, Genetic , Transgenes/physiology
20.
Hum Gene Ther ; 14(6): 497-507, 2003 Apr 10.
Article in English | MEDLINE | ID: mdl-12718761

ABSTRACT

The use of lentiviral vectors for basic research and potential future clinical applications requires methodologies that can accurately determine lentiviral titers and monitor viral transgene expression within target cells, beyond the context of reporter genes typically used for this purpose. Here we describe a quantitative RT-PCR (qRT-PCR) method that achieves both goals using primer sequences that are specific for the woodchuck hepatitis virus posttranscriptional regulatory element (WPRE), an enhancer contained in many retroviral vectors and that is incorporated in the 3' UTR of nascent transgene transcripts. Quantitation of titers of three recombinant lentiviruses, genetically identical except for the transgene, demonstrated consistent differences in titer that were likely due to transgene-associated toxicity in producer cells and target cells. Viruses encoding the tumor-associated antigens tyrosinase and neo-poly(A) polymerase yielded reproducibly lower titers than a virus encoding enhanced green fluorescent protein (GFP) at the viral RNA, integrated DNA, and transgene mRNA levels, as measured by WPRE qPCR. Furthermore, the magnitude of differences in expression of the three transgenes in transduced target cells could not have been predicted by measuring vector DNA integration events. Since transgene expression in target cells is the most common goal of lentiviral transduction, and since methods to quantify transgene expression on the protein level are not always readily available, qRT-PCR based on a nucleotide sequence included in the transcript provides a useful tool for titering novel recombinant lentiviruses.


Subject(s)
Genetic Vectors , Lentivirus/genetics , Reverse Transcriptase Polymerase Chain Reaction/methods , Transgenes , 3' Untranslated Regions/genetics , Cell Line , Cell Line, Transformed , Enhancer Elements, Genetic/genetics , Flow Cytometry , Gene Expression , Genes, Regulator/genetics , Green Fluorescent Proteins , HeLa Cells , Hepatitis Viruses/genetics , Humans , Luminescent Proteins/genetics , Luminescent Proteins/metabolism , RNA, Messenger/biosynthesis , Transduction, Genetic , Virion/genetics , Virus Integration
SELECTION OF CITATIONS
SEARCH DETAIL