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










Publication year range
1.
Eur J Ophthalmol ; : 11206721231201663, 2023 Sep 11.
Article in English | MEDLINE | ID: mdl-37697660

ABSTRACT

PURPOSE: To report medium-term visual acuity and refractive outcomes of patients with pseudoexfoliation implanted with toric or multifocal intraocular lenses (IOLs). METHODS: We retrospectively reviewed patients with pseudoexfoliation who had undergone phacoemulsification between 2016 and 2020 with at least 24 months follow-up. RESULTS: Mean follow-up was 44.17 ± 14.95 months. Toric IOLs were implanted in 48 eyes: mean uncorrected LogMAR visual acuity one month after surgery was 0.03 ± 0.09, decreasing to 0.08 ± 0.11 at the last visit (p = .01). Mean refractive cylinder was -0.12 ± 0.36 diopters one month after surgery and -0.25 ± 0.44 diopters at the last follow-up visit, p = .012. Multifocal IOLs were implanted in 42 patients: binocular uncorrected distance visual acuity was -0.02 ± 0.04 one month after surgery and 0.01 ± 0.05 at the last visit (p = .004); near acuity was 0.01 ± 0.03 and 0.04 ± 0.06 respectively (p = .001). In eyes with pseudoexfoliation, absolute spherical equivalent prediction error was 0.22 ± 0.20 diopters for toric and 0.21 ± 0.19 diopters for multifocal IOLs. One month after surgery 68.6% of eyes with toric IOLs and 74.2% of eyes with multifocal IOLs were within ±0.25 diopters of target spherical equivalent and 91.6% and 90.5% were within ±0.5 diopters, respectively. Spherical equivalent did not change significantly during follow-up for either group. CONCLUSION: Prediction error in eyes with pseudoexfoliation implanted with toric or multifocal IOLs was low and similar to values reported for normal eyes. Postoperative refractive cylinder with toric IOLs was low, with little change during follow-up. Visual function in patients receiving multifocal IOLs was excellent. Therefore, the implantation of these IOLs in eyes with pseudoexfoliation does not seem to cause medium-term problems.

2.
Eur J Cancer ; 144: 224-231, 2021 02.
Article in English | MEDLINE | ID: mdl-33373867

ABSTRACT

BACKGROUND: CDK4/6 inhibitors plus endocrine therapies are the current standard of care in the first-line treatment of HR+/HER2-negative metastatic breast cancer, but there are no well-established clinical or molecular predictive factors for patient response. In the era of personalised oncology, new approaches for developing predictive models of response are needed. MATERIALS AND METHODS: Data derived from the electronic health records (EHRs) of real-world patients with HR+/HER2-negative advanced breast cancer were used to develop predictive models for early and late progression to first-line treatment. Two machine learning approaches were used: a classic approach using a data set of manually extracted features from reviewed (EHR) patients, and a second approach using natural language processing (NLP) of free-text clinical notes recorded during medical visits. RESULTS: Of the 610 patients included, there were 473 (77.5%) progressions to first-line treatment, of which 126 (20.6%) occurred within the first 6 months. There were 152 patients (24.9%) who showed no disease progression before 28 months from the onset of first-line treatment. The best predictive model for early progression using the manually extracted dataset achieved an area under the curve (AUC) of 0.734 (95% CI 0.687-0.782). Using the NLP free-text processing approach, the best model obtained an AUC of 0.758 (95% CI 0.714-0.800). The best model to predict long responders using manually extracted data obtained an AUC of 0.669 (95% CI 0.608-0.730). With NLP free-text processing, the best model attained an AUC of 0.752 (95% CI 0.705-0.799). CONCLUSIONS: Using machine learning methods, we developed predictive models for early and late progression to first-line treatment of HR+/HER2-negative metastatic breast cancer, also finding that NLP-based machine learning models are slightly better than predictive models based on manually obtained data.


Subject(s)
Antineoplastic Combined Chemotherapy Protocols/therapeutic use , Breast Neoplasms/pathology , Machine Learning , Natural Language Processing , Receptor, ErbB-2/metabolism , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Adult , Aged , Aged, 80 and over , Breast Neoplasms/drug therapy , Breast Neoplasms/metabolism , Disease Progression , Electronic Health Records/statistics & numerical data , Female , Follow-Up Studies , Humans , Middle Aged , Prognosis , Retrospective Studies , Survival Rate , Young Adult
3.
Cells ; 9(10)2020 09 29.
Article in English | MEDLINE | ID: mdl-33003440

ABSTRACT

Podoplanin and CD44 are transmembrane glycoproteins involved in inflammation and cancer. In this paper, we report that podoplanin is coordinately expressed with the CD44 standard (CD44s) and variant (CD44v) isoforms in vivo-in hyperplastic skin after a pro-inflammatory stimulus with 12-O-tetradecanoylphorbol-13-acetate (TPA)-and in vitro-in cell lines representative of different stages of mouse-skin chemical carcinogenesis, as well as in human squamous carcinoma cell (SCC) lines. Moreover, we identify CD44v10 in the mouse-skin carcinogenesis model as the only CD44 variant isoform expressed in highly aggressive spindle carcinoma cell lines together with CD44s and podoplanin. We also characterized CD44v3-10, CD44v6-10 and CD44v8-10 as the major variant isoforms co-expressed with CD44s and podoplanin in human SCC cell lines. Immunofluorescence confocal microscopy experiments show that these CD44v isoforms colocalize with podoplanin at plasma membrane protrusions and cell-cell contacts of SCC cells, as previously reported for CD44s. Furthermore, CD44v isoforms colocalize with podoplanin in chemically induced mouse-skin SCCs in vivo. Co-immunoprecipitation experiments indicate that podoplanin physically binds to CD44v3-10, CD44v6-10 and CD44v8-10 isoforms, as well as to CD44s. Podoplanin-CD44 interaction is mediated by the transmembrane and cytosolic regions and is negatively modulated by glycosylation of the extracellular domain. These results point to a functional interplay of podoplanin with both CD44v and CD44s isoforms in SCCs and give insight into the regulation of the podoplanin-CD44 association.


Subject(s)
Carcinoma, Squamous Cell/metabolism , Hyaluronan Receptors/metabolism , Membrane Glycoproteins/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/metabolism , Carcinoma, Squamous Cell/genetics , Carcinoma, Squamous Cell/pathology , Cell Line, Tumor , Cell Membrane/metabolism , Cell Surface Extensions/metabolism , Humans , Hyaluronan Receptors/genetics , Membrane Glycoproteins/genetics , Mice , Phorbol Esters/pharmacology , Protein Binding , Protein Domains/genetics , Protein Isoforms
4.
Int J Mol Sci ; 20(8)2019 Apr 25.
Article in English | MEDLINE | ID: mdl-31027181

ABSTRACT

DUSP6/MKP3 is a dual-specific phosphatase that regulates extracellular regulated kinase ERK1/2 and ERK5 activity, with an increasingly recognized role as tumor suppressor. In silico studies from Gene expression Omnibus (GEO) and Cancer Genome atlas (TCGA) databases reveal poor prognosis in those Non-small cell lung cancer (NSCLC) patients with low expression levels of DUSP6. In agreement with these data, here we show that DUSP6 plays a major role in the regulation of cell migration, motility and tumor growth. We have found upregulation in the expression of several genes involved in epithelial to mesenchymal transition (EMT) in NSCLC-DUSP6 depleted cells. Data obtained in RNA-seq studies carried out in DUSP6 depleted cells identified EGFR, TGF-ß and WNT signaling pathways and several genes such as VAV3, RUNXR2, LEF1, FGFR2 whose expression is upregulated in these cells and therefore affecting cellular functions such as integrin mediated cell adhesion, focal adhesion and motility. Furthermore, EGF signaling pathway is activated via ERK5 and not ERK1/2 and TGF-ß via SMAD2/3 in DUSP6 depleted cells. In summary DUSP6 is a tumor suppressor in NSCLC and re-establishment of its expression may be a potential strategy to revert poor outcome in NSCLC patients.


Subject(s)
Carcinoma, Non-Small-Cell Lung/enzymology , Carcinoma, Non-Small-Cell Lung/genetics , Dual Specificity Phosphatase 6/genetics , Genes, Tumor Suppressor , Lung Neoplasms/enzymology , Lung Neoplasms/genetics , Actin Cytoskeleton/metabolism , Adenocarcinoma of Lung/enzymology , Adenocarcinoma of Lung/pathology , Adherens Junctions/metabolism , Animals , Carcinogenesis/genetics , Carcinogenesis/pathology , Carcinoma, Non-Small-Cell Lung/pathology , Cell Adhesion/genetics , Cell Line, Tumor , Cell Movement/genetics , Cell Shape/genetics , Disease Progression , Dual Specificity Phosphatase 6/metabolism , Focal Adhesions/metabolism , Gene Expression Regulation, Neoplastic , Humans , Kaplan-Meier Estimate , Lung Neoplasms/pathology , MAP Kinase Signaling System , Mice, Nude , Transforming Growth Factor beta/metabolism
5.
Int J Mol Sci ; 20(3)2019 Feb 06.
Article in English | MEDLINE | ID: mdl-30736372

ABSTRACT

Podoplanin is a small cell-surface mucin-like glycoprotein that plays a crucial role in the development of the alveoli, heart, and lymphatic vascular system. Emerging evidence indicates that it is also involved in the control of mammary stem-cell activity and biogenesis of platelets in the bone marrow, and exerts an important function in the immune response. Podoplanin expression is upregulated in different cell types, including fibroblasts, macrophages, T helper cells, and epithelial cells, during inflammation and cancer, where it plays important roles. Podoplanin is implicated in chronic inflammatory diseases, such as psoriasis, multiple sclerosis, and rheumatoid arthritis, promotes inflammation-driven and cancer-associated thrombosis, and stimulates cancer cell invasion and metastasis through a variety of strategies. To accomplish its biological functions, podoplanin must interact with other proteins located in the same cell or in neighbor cells. The binding of podoplanin to its ligands leads to modulation of signaling pathways that regulate proliferation, contractility, migration, epithelial⁻mesenchymal transition, and remodeling of the extracellular matrix. In this review, we describe the diverse roles of podoplanin in inflammation and cancer, depict the protein ligands of podoplanin identified so far, and discuss the mechanistic basis for the involvement of podoplanin in all these processes.


Subject(s)
Inflammation/etiology , Inflammation/metabolism , Membrane Glycoproteins/genetics , Membrane Glycoproteins/metabolism , Neoplasms/etiology , Neoplasms/metabolism , Animals , Carrier Proteins , Cell Differentiation/genetics , Cell Transformation, Neoplastic/genetics , Cell Transformation, Neoplastic/metabolism , Disease Susceptibility , Embryonic Development/genetics , Gene Expression Regulation , Humans , Immunomodulation , Lymphangiogenesis/genetics , Membrane Glycoproteins/chemistry , Protein Binding , Signal Transduction , Structure-Activity Relationship
6.
Sci Rep ; 7(1): 6276, 2017 07 24.
Article in English | MEDLINE | ID: mdl-28740236

ABSTRACT

Qa-2 is believed to mediate a protective immune response against cancer; however, little is known about the role of Qa-2 in tumorigenesis. Here, we used 4T1 breast cancer cells to study the involvement of Qa-2 in tumor progression in a syngeneic host. Qa-2 expression was reduced during in vivo tumor growth and in cell lines derived from 4T1-induced tumors. Tumor-derived cells elicited an epithelial-mesenchymal transition associated with upregulation of Zeb1 and Twist1/2 and enhanced tumor initiating and invasive capacities. Furthermore, these cells showed increased stem characteristics, as demonstrated by upregulation of Hes1, Sox2 and Oct3/4, and enrichment of CD44high/CD24median/low cells. Remarkably, Qa-2 cell-surface expression was excluded from the CD44high/CD24median/low subpopulation. Tumor-derived cells showed increased Src activity, and treatment of these cells with the Src kinase inhibitor PP2 enhanced Qa-2 but reduced Sox2 and CD44high/CD24median/low expression levels, suggesting that Src signaling, while positively associated with stemness, negatively regulates Qa-2 expression in breast cancer. Finally, overexpression of the Qa-2 family member Q7 on the cell surface slowed down in vivo tumor growth and reduced the metastatic potential of 4T1 cells. These results suggest an anti-malignant role for Qa-2 in breast cancer development, which appears to be absent from cancer stem cells.


Subject(s)
Breast Neoplasms/pathology , Epithelial-Mesenchymal Transition , Histocompatibility Antigens Class I/metabolism , Neoplastic Stem Cells/pathology , Animals , Apoptosis , Breast Neoplasms/metabolism , Carcinogenesis , Cell Movement , Cell Proliferation , Female , Humans , Mice , Mice, Inbred BALB C , Neoplastic Stem Cells/metabolism , Tumor Cells, Cultured , Xenograft Model Antitumor Assays
7.
Sci Rep ; 6: 27544, 2016 06 08.
Article in English | MEDLINE | ID: mdl-27270881

ABSTRACT

Antithrombin is a key inhibitor of the coagulation cascade, but it may also function as an anti-inflammatory, anti-angiogenic, anti-viral and anti-apoptotic protein. Here, we report a novel function of antithrombin as a modulator of tumor cell migration and invasion. Antithrombin inhibited enteropeptidase on the membrane surface of HT-29, A549 and U-87 MG cells. The inhibitory process required the activation of antithrombin by heparin, and the reactive center loop and the heparin binding domain were essential. Surprisingly, antithrombin non-covalently inhibited enteropeptidase, revealing a novel mechanism of inhibition for this serpin. Moreover, as a consequence of this inhibition, antithrombin was cleaved, resulting in a molecule with anti-angiogenic properties that reduced vessel-like formation of endothelial cells. The addition of antithrombin and heparin to U-87 MG and A549 cells reduced motility in wound healing assays, inhibited the invasion in transwell assays and the degradation of a gelatin matrix mediated by invadopodia. These processes were controlled by enteropeptidase, as demonstrated by RNA interference experiments. Carcinoma cell xenografts in nude mice showed in vivo co-localization of enteropeptidase and antithrombin. Finally, treatment with heparin reduced experimental metastasis induced by HT29 cells in vivo. In conclusion, the inhibition of enteropeptidase by antithrombin may have a double anti-tumor effect through inhibiting a protease involved in metastasis and generating an anti-angiogenic molecule.


Subject(s)
Antithrombins/administration & dosage , Enteropeptidase/genetics , Neoplasms/drug therapy , Neovascularization, Pathologic/drug therapy , A549 Cells , Angiogenesis Inhibitors/administration & dosage , Animals , Cell Movement/drug effects , Endothelial Cells/drug effects , Endothelial Cells/pathology , HT29 Cells , Humans , Mice , Neoplasm Invasiveness/genetics , Neoplasm Invasiveness/pathology , Neoplasms/genetics , Neoplasms/pathology , Neovascularization, Pathologic/genetics , Neovascularization, Pathologic/pathology , Protein Binding , Protein Conformation
8.
Oncotarget ; 7(13): 16070-89, 2016 Mar 29.
Article in English | MEDLINE | ID: mdl-26893367

ABSTRACT

Podoplanin (PDPN) is a transmembrane glycoprotein that plays crucial roles in embryonic development, the immune response, and malignant progression. Here, we report that cells ectopically or endogenously expressing PDPN release extracellular vesicles (EVs) that contain PDPN mRNA and protein. PDPN incorporates into membrane shed microvesicles (MVs) and endosomal-derived exosomes (EXOs), where it was found to colocalize with the canonical EV marker CD63 by immunoelectron microscopy. We have previously found that expression of PDPN in MDCK cells induces an epithelial-mesenchymal transition (EMT). Proteomic profiling of MDCK-PDPN cells compared to control cells shows that PDPN-induced EMT is associated with upregulation of oncogenic proteins and diminished expression of tumor suppressors. Proteomic analysis of exosomes reveals that MDCK-PDPN EXOs were enriched in protein cargos involved in cell adhesion, cytoskeletal remodeling, signal transduction and, importantly, intracellular trafficking and EV biogenesis. Indeed, expression of PDPN in MDCK cells stimulated both EXO and MV production, while knockdown of endogenous PDPN in human HN5 squamous carcinoma cells reduced EXO production and inhibited tumorigenesis. EXOs released from MDCK-PDPN and control cells both stimulated in vitro angiogenesis, but only EXOs containing PDPN were shown to promote lymphatic vessel formation. This effect was mediated by PDPN on the surface of EXOs, as demonstrated by a neutralizing specific monoclonal antibody. These results contribute to our understanding of PDPN-induced EMT in association to tumor progression, and suggest an important role for PDPN in EV biogenesis and/or release and for PDPN-EXOs in modulating lymphangiogenesis.


Subject(s)
Epithelial-Mesenchymal Transition/physiology , Exosomes/metabolism , Extracellular Vesicles/metabolism , Lymphangiogenesis/physiology , Membrane Glycoproteins/metabolism , Animals , Cell Line, Tumor , Cell Transformation, Neoplastic/metabolism , Dogs , Heterografts , Humans , Madin Darby Canine Kidney Cells , Mice , Mice, Inbred BALB C , Neoplasms, Experimental/metabolism , Neoplasms, Experimental/pathology
9.
Int Rev Cell Mol Biol ; 317: 185-239, 2015.
Article in English | MEDLINE | ID: mdl-26008786

ABSTRACT

Podoplanin is a small mucin-like transmembrane protein expressed in several adult tissues and with an important role during embryogenesis. It is needed for the proper development of kidneys and lungs as well as accurate formation of the lymphatic vascular system. In addition, it is involved in the physiology of the immune system. A wide variety of tumors express podoplanin, both in the malignant cells and in the stroma. Although there are exceptions, the presence of podoplanin results in poor prognosis. The main consequence of forced podoplanin expression in established and tumor-derived cell lines is an increase in cell migration and, eventually, the triggering of an epithelial-mesenchymal transition, whereby cells acquire a fibroblastoid phenotype and increased motility. We will examine the current status of the role of podoplanin in the induction of epithelial-mesenchymal transition as well as the different interactions that lead to this program.


Subject(s)
Cell Movement , Epithelial-Mesenchymal Transition , Membrane Glycoproteins/metabolism , Neoplasms/metabolism , Neoplasms/pathology , Adult , Animals , Humans
10.
J Natl Cancer Inst ; 107(6): djv077, 2015 Jun.
Article in English | MEDLINE | ID: mdl-25855725

ABSTRACT

BACKGROUND: Pharmacological activation of cannabinoid receptors elicits antitumoral responses in different cancer models. However, the biological role of these receptors in tumor physio-pathology is still unknown. METHODS: We analyzed CB2 cannabinoid receptor protein expression in two series of 166 and 483 breast tumor samples operated in the University Hospitals of Kiel, Tübingen, and Freiburg between 1997 and 2010 and CB2 mRNA expression in previously published DNA microarray datasets. The role of CB2 in oncogenesis was studied by generating a mouse line that expresses the human V-Erb-B2 Avian Erythroblastic Leukemia Viral Oncogene Homolog 2 (HER2) rat ortholog (neu) and lacks CB2 and by a variety of biochemical and cell biology approaches in human breast cancer cells in culture and in vivo, upon modulation of CB2 expression by si/shRNAs and overexpression plasmids. CB2-HER2 molecular interaction was studied by colocalization, coimmunoprecipitation, and proximity ligation assays. Statistical tests were two-sided. RESULTS: We show an association between elevated CB2 expression in HER2+ breast tumors and poor patient prognosis (decreased overall survival, hazard ratio [HR] = 0.29, 95% confidence interval [CI] = 0.09 to 0.71, P = .009) and higher probability to suffer local recurrence (HR = 0.09, 95% CI = 0.049 to 0.54, P = .003) and to develop distant metastases (HR = 0.33, 95% CI = 0.13 to 0.75, P = .009). We also demonstrate that genetic inactivation of CB2 impairs tumor generation and progression in MMTV-neu mice. Moreover, we show that HER2 upregulates CB2 expression by activating the transcription factor ELK1 via the ERK cascade and that an increased CB2 expression activates the HER2 pro-oncogenic signaling at the level of the tyrosine kinase c-SRC. Finally, we show HER2 and CB2 form heteromers in cancer cells. CONCLUSIONS: Our findings reveal an unprecedented role of CB2 as a pivotal regulator of HER2 pro-oncogenic signaling in breast cancer, and they suggest that CB2 may be a biomarker with prognostic value in these tumors.


Subject(s)
Biomarkers, Tumor/metabolism , Breast Neoplasms/metabolism , Receptor, Cannabinoid, CB2/metabolism , Receptor, ErbB-2/metabolism , Signal Transduction , Animals , Cell Line, Tumor , Female , Gene Expression Regulation, Neoplastic , Germany , Humans , Immunohistochemistry , Immunoprecipitation , Kaplan-Meier Estimate , Mice , Prognosis , RNA, Messenger/metabolism , Receptor, Cannabinoid, CB2/genetics , Receptor, ErbB-2/genetics , Receptors, Estrogen/metabolism , Receptors, Progesterone/metabolism , Tissue Array Analysis , Transcription, Genetic
11.
Carcinogenesis ; 36(2): 212-22, 2015 Feb.
Article in English | MEDLINE | ID: mdl-25503931

ABSTRACT

Increased levels of soluble endoglin (Sol-Eng) correlate with poor outcome in human cancer. We have previously shown that shedding of membrane endoglin, and concomitant release of Sol-Eng is a late event in chemical mouse skin carcinogenesis associated with the development of undifferentiated spindle cell carcinomas (SpCCs). In this report, we show that mouse skin SpCCs exhibit a high expression of hepatocyte growth factor (HGF) and an elevated ratio of its active tyrosine kinase receptor Met versus total Met levels. We have evaluated the effect of Sol-Eng in spindle carcinoma cells by transfection of a cDNA encoding most of the endoglin ectodomain or by using purified recombinant Sol-Eng. We found that Sol-Eng inhibited both mitogen-activated protein kinase (MAPK) activity and cell growth in vitro and in vivo. Sol-Eng also blocked MAPK activation by transforming growth factor-ß1 (TGF-ß1) and impaired both basal and HGF-induced activation of Met and downstream MAPK. Moreover, Sol-Eng strongly reduced basal and HGF-stimulated spindle cell migration and invasion. Both Sol-Eng and full-length endoglin were shown to interact with Met by coimmunoprecipitation experiments. However, full-length endoglin expressed at the plasma membrane of spindle carcinoma cells had no effect on Met signaling activity, and was unable to inhibit HGF-induced cell migration/invasion. These results point to a paradoxical suppressor role for Sol-Eng in carcinogenesis.


Subject(s)
Antigens, CD/metabolism , Carcinogenesis/metabolism , Hepatocyte Growth Factor/biosynthesis , Proto-Oncogene Proteins c-met/antagonists & inhibitors , Receptors, Cell Surface/metabolism , Sarcoma/metabolism , Skin Neoplasms/metabolism , 9,10-Dimethyl-1,2-benzanthracene/pharmacology , Animals , Antigens, CD/genetics , Carcinogenesis/pathology , Cell Movement/genetics , Cell Proliferation/genetics , DNA, Complementary/genetics , Endoglin , Enzyme Activation , Female , HEK293 Cells , Humans , MAP Kinase Signaling System , Mice , Mice, Nude , Mitogen-Activated Protein Kinases/antagonists & inhibitors , Prognosis , Receptors, Cell Surface/genetics , Sarcoma/pathology , Skin Neoplasms/pathology , Tetradecanoylphorbol Acetate/pharmacology , Transfection , Transforming Growth Factor beta1/antagonists & inhibitors , Tumor Cells, Cultured
12.
Int J Biochem Cell Biol ; 50: 47-54, 2014 May.
Article in English | MEDLINE | ID: mdl-24508783

ABSTRACT

Podosomes are integrin-based adhesions fundamental for stabilisation of the leading lamellae in migrating dendritic cells (DCs) and for extracellular matrix (ECM) degradation. We have previously shown that soluble factors and chemokines such as SDF 1-a trigger podosome initiation whereas integrin ligands promote podosome maturation and stability in DCs. The exact intracellular signalling pathways that regulate the sequential organisation of podosomal components in response to extracellular cues remain largely undetermined. The Wiskott Aldrich Syndrome Protein (WASP) mediates actin polymerisation and the initial recruitment of integrins and associated proteins in a circular configuration surrounding the core of filamentous actin (F-actin) during podosome initiation. We have now identified integrin linked kinase (ILK) surrounding the podosomal actin core. We report that DC polarisation in response to chemokines and the assembly of actin cores during podosome initiation require PI3K-dependent clustering of the Wiskott Aldrich Syndrome Protein (WASP) in puncta independently of ILK. ILK is essential for the clustering of integrins and associated proteins leading to podosome maturation and stability that are required for degradation of the subjacent extracellular matrix and the invasive motility of DCs across connective tissue barriers. We conclude that WASP regulates DCs polarisation for migration and initiation of actin polymerisation downstream of PI3K in nascent podosomes. Subsequently, ILK mediates the accumulation of integrin-associated proteins during podosome maturation and stability for efficient degradation of the subjacent ECM during the invasive migration of DCs.


Subject(s)
Dendritic Cells/cytology , Dendritic Cells/enzymology , Protein Serine-Threonine Kinases/metabolism , Animals , Cell Membrane Structures/enzymology , Cell Movement/physiology , Dendritic Cells/metabolism , Extracellular Matrix/metabolism , Humans , Male , Mice , Mice, Inbred C57BL , Phosphatidylinositol 3-Kinases/metabolism , Transfection , Wiskott-Aldrich Syndrome Protein/metabolism
13.
Int J Biochem Cell Biol ; 46: 68-75, 2014 Jan.
Article in English | MEDLINE | ID: mdl-24275092

ABSTRACT

Podoplanin (PDPN) is a mucin-like transmembrane glycoprotein that plays an important role in development and cancer. Here, we provide evidence that the intracellular domain (ICD) of podoplanin is released into the cytosol following a sequential proteolytic processing by a metalloprotease and γ-secretase. Western blotting and cell fractionation studies revealed that HEK293T and MDCK cells transfected with an eGFP-tagged podoplanin construct (PDPNeGFP, 50-63kDa) constitutively express two C-terminal fragments (CTFs): a ∼33kDa membrane-bound PCTF33, and a ∼29kDa cytosolic podoplanin ICD (PICD). While pharmacological inhibition of metalloproteases reduced the expression of PCTF33, treatment of cells with γ-secretase inhibitors resulted in enhanced PCTF33 levels. PCTF33 processing by γ-secretase depends on presenilin-1 (PS1) function: cells expressing a dominant negative form of PS1 (PS1 D385N), and mouse embryonic fibroblasts (MEFs) genetically deficient in PS1, but not in PS2, show higher levels of PCTF33 expression with respect to wild-type MEFs. Furthermore, transfection of PS1 deficient MEFs with wild-type PS1 (PS1 wt) decreased PCTF33 levels. N-terminal amino acid sequencing of the affinity purified PICD revealed that the γ-secretase cleavage site was located between valines 150 and 151, but these residues are not critical for proteolysis. We found that podoplanin CTFs are also generated in cells expressing podoplanin mutants harboring heterologous transmembrane regions. Taken together, these results indicate that podoplanin is a novel substrate for PS1/γ-secretase.


Subject(s)
Amyloid Precursor Protein Secretases/metabolism , Membrane Glycoproteins/metabolism , Presenilin-1/metabolism , Amyloid Precursor Protein Secretases/genetics , Animals , Dogs , HEK293 Cells , Humans , Madin Darby Canine Kidney Cells , Mice , Mice, Transgenic , Presenilin-1/genetics , Transfection
14.
J Invest Dermatol ; 134(1): 247-255, 2014 Jan.
Article in English | MEDLINE | ID: mdl-23765132

ABSTRACT

Endoglin (Eng) is a transmembrane glycoprotein that is mainly expressed in endothelial cells, but it is also present in the epidermis and skin appendages. To address the role of Eng in cutaneous wound healing, we compared the kinetics of reepithelialization in Eng heterozygous null (Eng(+/-)) mice and their normal littermates (Eng(+/+)) following skin wounds. The wound area was significantly larger in Eng(+/-) than in Eng(+/+) mice from 2 to 8 days after injury; overall wound closure was delayed by 1 to 2 days. In Eng(+/-) mice, keratinocytes at the wound edges exhibited impaired proliferation but were more migratory, as shown by their elongated morphology and increased keratin 17 expression. Inhibition of nitric oxide (NO) synthesis delayed healing in Eng(+/+) but not in Eng(+/-) mice. Administration of the NO donor LA-803 accelerated wound closure in Eng(+/-) mice, with no effect on normal littermates. The acute stimulation with 12-O-tetradecanoylphorbol-13-acetate (TPA) enhanced Eng expression in mouse epidermal keratinocytes in vivo and in vitro associated with hyperproliferation. Similarly, the skin of Eng(+/-) mice failed to mount a hyperplastic response to acute stimulation with TPA. These results demonstrate an important involvement of Eng in wound healing that is associated with NO bioavailability.


Subject(s)
Epidermis/injuries , Intracellular Signaling Peptides and Proteins/genetics , Nitric Oxide/metabolism , Wound Healing/physiology , Age Factors , Animals , Carcinogens/pharmacology , Cell Proliferation , Endoglin , Epidermis/metabolism , Epidermis/pathology , Epithelial Cells/drug effects , Epithelial Cells/metabolism , Female , Heterozygote , Hyperplasia , Intracellular Signaling Peptides and Proteins/metabolism , Mice , Mice, Inbred C57BL , Mice, Knockout , Tetradecanoylphorbol Acetate/pharmacology
15.
Int J Biochem Cell Biol ; 43(6): 886-96, 2011 Jun.
Article in English | MEDLINE | ID: mdl-21376833

ABSTRACT

Podoplanin is a transmembrane glycoprotein that is upregulated in cancer and was reported to induce an epithelial-mesenchymal transition (EMT) in MDCK cells. The promotion of EMT was dependent on podoplanin binding to ERM (ezrin, radixin, moesin) proteins through its cytoplasmic (CT) domain, which led to RhoA-associated kinase (ROCK)-dependent ERM phosphorylation. Using detergent-resistant membrane (DRM) assays, as well as transmembrane (TM) interactions and ganglioside GM1 binding, we present evidence supporting the localization of podoplanin in raft platforms important for cell signalling. Podoplanin mutant constructs harbouring a heterologous TM region or lacking the CT tail were unable to associate with DRMs, stimulate ERM phosphorylation and promote EMT or cell migration. Similar effects were observed upon disruption of a GXXXG motif within the TM domain, which is involved in podoplanin self-assembly. In contrast, deletion of the extracellular (EC) domain did not affect podoplanin DRM association. Together, these data suggest that both the CT and TM domains are required for podoplanin localization in raft platforms, and that this association appears to be necessary for podoplanin-mediated EMT and cell migration.


Subject(s)
Epithelial-Mesenchymal Transition , Membrane Glycoproteins/metabolism , Membrane Microdomains/metabolism , Amino Acid Motifs/genetics , Animals , Cell Line , Dogs , Epithelial-Mesenchymal Transition/genetics , Ganglioside Galactosyltransferase/metabolism , Humans , Membrane Glycoproteins/genetics , Mutation/genetics , Protein Binding , Protein Structure, Tertiary/genetics , Protein Transport/genetics , Signal Transduction , rho-Associated Kinases/metabolism
16.
Mol Biol Cell ; 21(24): 4387-99, 2010 Dec.
Article in English | MEDLINE | ID: mdl-20962267

ABSTRACT

Podoplanin is a transmembrane glycoprotein up-regulated in different human tumors, especially those derived from squamous stratified epithelia (SCCs). Its expression in tumor cells is linked to increased cell migration and invasiveness; however, the mechanisms underlying this process remain poorly understood. Here we report that CD44, the major hyaluronan (HA) receptor, is a novel partner for podoplanin. Expression of the CD44 standard isoform (CD44s) is coordinately up-regulated together with that of podoplanin during progression to highly aggressive SCCs in a mouse skin model of carcinogenesis, and during epithelial-mesenchymal transition (EMT). In carcinoma cells, CD44 and podoplanin colocalize at cell surface protrusions. Moreover, CD44 recruitment promoted by HA-coated beads or cross-linking with a specific CD44 antibody induced corecruitment of podoplanin. Podoplanin-CD44s interaction was demonstrated both by coimmunoprecipitation experiments and, in vivo, by fluorescence resonance energy transfer/fluorescence lifetime imaging microscopy (FRET/FLIM), the later confirming its association on the plasma membrane of cells with a migratory phenotype. Importantly, we also show that podoplanin promotes directional persistence of motility in epithelial cells, a feature that requires CD44, and that both molecules cooperate to promote directional migration in SCC cells. Our results support a role for CD44-podoplanin interaction in driving tumor cell migration during malignancy.


Subject(s)
Carcinoma, Squamous Cell/pathology , Cell Movement , Hyaluronan Receptors/metabolism , Membrane Glycoproteins/metabolism , Skin Neoplasms/pathology , Animals , Cell Adhesion , Cells, Cultured , Epithelial-Mesenchymal Transition , Fluorescence Resonance Energy Transfer , Humans , Hyaluronan Receptors/genetics , Hyaluronic Acid/metabolism , Membrane Glycoproteins/genetics , Mice , Microscopy, Fluorescence , Neoplasm Invasiveness , Protein Isoforms/genetics , Protein Isoforms/metabolism , Up-Regulation
17.
Carcinogenesis ; 30(8): 1459-68, 2009 Aug.
Article in English | MEDLINE | ID: mdl-19502595

ABSTRACT

Vitamin D receptor (VDR) mediates the antitumoral action of the active vitamin D metabolite 1alpha,25-dihydroxyvitamin D(3) (1,25(OH)(2)D(3)). VDR expression is lost during colon cancer progression causing unresponsiveness to 1,25(OH)(2)D(3) and its analogs. Previously, Snail1, an inducer of epithelial-to-mesenchymal transition (EMT), was reported to inhibit VDR expression. Here, we show that Snail2/Slug, but not other EMT inducers such as Zeb1, Zeb2, E47 or Twist1, represses VDR gene promoter. Moreover, Snail2 and Snail1 show additive repressing effect on VDR promoter. Snail2 inhibits VDR RNA and protein and blocks the induction of E-cadherin and an adhesive phenotype by 1,25(OH)(2)D(3). Snail2 reduces the ligand-induced VDR transcriptional activation of a consensus response element and of the CYP24 promoter. Concordantly, Snail2 inhibits the induction of CYP24 RNA and p21(CIP1), filamin A and vinculin proteins and the repression of c-MYC by 1,25(OH)(2)D(3). Additionally, Snail2 abrogates beta-catenin nuclear export and the antagonism of the transcriptional activity of beta-catenin-T-cell factor complexes by 1,25(OH)(2)D(3). SNAI2 expression is upregulated in 58% of colorectal tumors and correlates inversely with that of VDR. However, VDR downregulation is higher in tumors coexpressing SNAI2 and SNAI1 than in those expressing only one of these genes. Together, these data indicate that Snail2 and Snail1 cooperate for VDR repression in colon cancer.


Subject(s)
Adenocarcinoma/metabolism , Colorectal Neoplasms/metabolism , Colorectal Neoplasms/pathology , Receptors, Calcitriol/genetics , Transcription Factors/metabolism , Transcription, Genetic , Adenocarcinoma/genetics , Adenocarcinoma/secondary , Blotting, Western , Cadherins/genetics , Cadherins/metabolism , Colorectal Neoplasms/genetics , Fluorescent Antibody Technique , Humans , Immunoenzyme Techniques , Nuclear Proteins/genetics , Nuclear Proteins/metabolism , Promoter Regions, Genetic , RNA, Messenger/genetics , RNA, Messenger/metabolism , Receptors, Calcitriol/antagonists & inhibitors , Receptors, Calcitriol/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Signal Transduction , Snail Family Transcription Factors , Transcription Factors/genetics , Twist-Related Protein 1/genetics , Twist-Related Protein 1/metabolism , Vitamin D/analogs & derivatives , Vitamin D/pharmacology , Wnt Proteins/genetics , Wnt Proteins/metabolism , beta Catenin/genetics , beta Catenin/metabolism
18.
Int J Biochem Cell Biol ; 41(6): 1421-9, 2009 Jun.
Article in English | MEDLINE | ID: mdl-19146981

ABSTRACT

Podoplanin/PA2.26 antigen is a small transmembrane mucin expressed in different types of cancer where it is associated with increased cell migration, invasiveness and metastasis. Little is known about the mechanisms that control podoplanin expression. Here, we show that podoplanin synthesis can be controlled at different levels. We analyzed podoplanin expression in a wide panel of tumour cell lines. The podoplanin gene (PDPN) is transcribed in cells derived from sarcomas, embryonal carcinomas, squamous cell carcinomas and endometrial tumours, while cell lines derived from colon, pancreatic, ovarian and ductal breast carcinomas do not express PDPN transcripts. PDPN is expressed as two mRNAs of approximately 2.7 and approximately 0.9 kb, both of which contain the coding sequence and arise by alternative polyadenylation. Strikingly, in most of the cell lines where PDPN transcripts were found, no podoplanin or only very low levels of the protein could be detected in Western blot. Treatment of several of these cell lines with the calpain inhibitor calpeptin resulted in podoplanin accumulation, whereas lactacystin, a specific inhibitor of the proteasome, had no effect. In vitro experiments showed that podoplanin is a substrate of calpain-1. These results indicate that at least in some tumour cells absence or reduced podoplanin protein levels are due to post-translational calpain-mediated proteolysis. We also report in this article the identification of a novel podoplanin isoform that originates by alternative splicing and differs from the standard form in lacking two cytoplasmic residues (YS). YS dipeptide is highly conserved across species, suggesting that it might be functionally relevant.


Subject(s)
Calpain/metabolism , Membrane Glycoproteins/biosynthesis , Neoplasms/metabolism , Alternative Splicing , Amino Acid Sequence , Base Sequence , Calpain/genetics , Cell Line, Tumor , Dipeptides/pharmacology , Gene Expression , Glycoproteins/pharmacology , Humans , Membrane Glycoproteins/genetics , Molecular Sequence Data , Neoplasms/genetics , Protein Processing, Post-Translational , RNA, Messenger/biosynthesis , RNA, Messenger/genetics , Transcriptional Activation
19.
J Cell Sci ; 119(Pt 21): 4541-53, 2006 Nov 01.
Article in English | MEDLINE | ID: mdl-17046996

ABSTRACT

Podoplanin is a small membrane mucin expressed in tumors associated with malignant progression. It is enriched at cell-surface protrusions where it colocalizes with members of the ERM (ezrin, radixin, moesin) protein family. Here, we found that human podoplanin directly interacts with ezrin (and moesin) in vitro and in vivo through a cluster of basic amino acids within its cytoplasmic tail, mainly through a juxtamembrane dipeptide RK. Podoplanin induced an epithelial-mesenchymal transition in MDCK cells linked to the activation of RhoA and increased cell migration and invasiveness. Fluorescence time-lapse video observations in migrating cells indicate that podoplanin might be involved in ruffling activity as well as in retractive processes. By using mutant podoplanin constructs fused to green fluorescent protein we show that association of the cytoplasmic tail with ERM proteins is required for upregulation of RhoA activity and epithelial-mesenchymal transition. Furthermore, expression of either a dominant-negative truncated variant of ezrin or a dominant-negative mutant form of RhoA blocked podoplanin-induced RhoA activation and epithelial-mesenchymal transition. These results provide a mechanistic basis to understand the role of podoplanin in cell migration or invasiveness.


Subject(s)
Cytoskeletal Proteins/metabolism , Epithelial Cells/pathology , Membrane Glycoproteins/metabolism , Membrane Proteins/metabolism , Mesoderm/pathology , Microfilament Proteins/metabolism , rhoA GTP-Binding Protein/metabolism , Animals , Blotting, Western , Cell Line , Cell Movement , Dogs , Epithelial Cells/metabolism , Fluorescent Antibody Technique , HeLa Cells , Humans , Kidney/metabolism , Membrane Glycoproteins/genetics , Mesoderm/metabolism , Reverse Transcriptase Polymerase Chain Reaction , Wound Healing , rhoA GTP-Binding Protein/genetics
20.
Int J Cancer ; 113(6): 899-910, 2005 Mar 01.
Article in English | MEDLINE | ID: mdl-15515019

ABSTRACT

We report the full cDNA sequence encoding the human homologue of murine PA2.26 (T1alpha-2, podoplanin), a small mucin-type transmembrane glycoprotein originally identified as a cell-surface antigen induced in keratinocytes during mouse skin carcinogenesis. The human PA2.26 gene is expressed as 2 transcripts of 0.9 and 2.7 kb in several normal tissues, such as the placenta, skeletal muscle, heart and lung. Using a specific polyclonal antibody raised against a synthetic peptide of the protein ectodomain, PA2.26 was immunohistochemically detected in about 25% (15/61) of human early oral squamous cell carcinomas. PA2.26 distribution in the tumours was heterogeneous and often restricted to the invasive front. Double immunofluorescence and confocal microscopy analysis showed that PA2.26 colocalized with the membrane cytoskeleton linker ezrin at the surface of tumour cells and that its presence in vivo was associated with downregulation of membrane E-cadherin protein expression. Ectopic expression of human PA2.26 in HeLa carcinoma cells and immortalized HaCaT keratinocytes promoted a redistribution of ezrin to the cell edges, the formation of cell-surface protrusions and reduced Ca(2+)-dependent cell-cell adhesiveness. These results point to PA2.26 as a novel biomarker for oral squamous cell carcinomas that might be involved in migration/invasion.


Subject(s)
Antigens, Surface/genetics , Carcinoma, Squamous Cell/genetics , Membrane Glycoproteins/genetics , Mouth Neoplasms/genetics , Amino Acid Sequence , Animals , Base Sequence , Carcinoma, Squamous Cell/pathology , Female , Humans , Mice , Molecular Sequence Data , Mouth Neoplasms/pathology , Neoplasm Invasiveness , Neoplasm Staging , Organ Specificity , Pregnancy , Protein Biosynthesis , RNA, Messenger/genetics , Transcription, Genetic
SELECTION OF CITATIONS
SEARCH DETAIL
...